Skip to main content
BMC Cancer logoLink to BMC Cancer
. 2015 Mar 11;15:117. doi: 10.1186/s12885-015-1101-8

Prediction of resistance to chemotherapy in ovarian cancer: a systematic review

Katherine L Lloyd 1,, Ian A Cree 2, Richard S Savage 2,3
PMCID: PMC4371880  PMID: 25886033

Abstract

Background

Patient response to chemotherapy for ovarian cancer is extremely heterogeneous and there are currently no tools to aid the prediction of sensitivity or resistance to chemotherapy and allow treatment stratification. Such a tool could greatly improve patient survival by identifying the most appropriate treatment on a patient-specific basis.

Methods

PubMed was searched for studies predicting response or resistance to chemotherapy using gene expression measurements of human tissue in ovarian cancer.

Results

42 studies were identified and both the data collection and modelling methods were compared. The majority of studies utilised fresh-frozen or formalin-fixed paraffin-embedded tissue. Modelling techniques varied, the most popular being Cox proportional hazards regression and hierarchical clustering which were used by 17 and 11 studies respectively. The gene signatures identified by the various studies were not consistent, with very few genes being identified by more than two studies. Patient cohorts were often noted to be heterogeneous with respect to chemotherapy treatment undergone by patients.

Conclusions

A clinically applicable gene signature capable of predicting patient response to chemotherapy has not yet been identified. Research into a predictive, as opposed to prognostic, model could be highly beneficial and aid the identification of the most suitable treatment for patients.

Electronic supplementary material

The online version of this article (doi:10.1186/s12885-015-1101-8) contains supplementary material, which is available to authorized users.

Keywords: Ovarian cancer, Chemoresistance, Predictive model, Statistical modelling

Background

Ovarian cancer is the fifth most common cancer in women in the UK and accounted for 4% of cancer diagnoses in women between 2008 and 2010 [1]. Worryingly, it was also responsible for 6% of cancer-related deaths in women over the same time period [1] and the five-year survival of women diagnosed with ovarian cancer between 2005 and 2009 was 42% [2]. It has been observed that although 40%-60% of patients achieve complete clinical response to first-line chemotherapy treatment [3], around 50% of these patients relapse within 5 years [4] and only 10%-15% of patients presenting with advanced stage disease achieve long-term remission [5]. It is thought that the high relapse rate is at least in part due to resistance to chemotherapy, which may be inherent or acquired by altered gene expression [6].

For ovarian cancer in the UK, the standard of care for first-line chemotherapy treatment recommended by the National Institute for Health and Care Excellence is ‘paclitaxel in combination with a platinum-based compound or platinum-based therapy alone’ [7]. This uniform approach ignores the complexity of ovarian cancer histologic types, particularly as there is evidence to suggest differences in response [8]. Winter et al. [9] investigated the survival of patients following paclitaxel and platinum chemotherapy and found histology to be a significant predictor of overall survival in multivariate Cox proportional hazards regression.

Improvement in survival has also been poor in ovarian cancer. Between 1971 and 2007 there was a 38% increase in relative 10-year survival in breast cancer, whereas the increase in ovarian cancer was 17% [10]. This difference in progress is likely to be due, at least in part, to the lack of tools with which to predict chemotherapy response in ovarian cancer.

Gene expression based tools for the prediction of patient prognosis after surgery or chemotherapy are currently available for some cancers. For example, MammaPrint®; uses the expression of 70 genes to predict the likelihood of metastasis in breast cancer [11]. Similarly, the Oncotype DX®; assay uses the expression of a panel of 21 genes to predict recurrence after treatment of breast cancer [12]. The Oncotype DX assay is also available for colon [13] and prostate cancers [14]. The development of a similar tool for ovarian cancer could greatly improve patient prognosis and quality of life by guiding chemotherapy choices. The prediction of cancer prognosis using gene signatures is a popular research field, within which a wide variety of approaches have been considered. Popular RNA or protein expression measurement techniques include cDNA hybridisation microarrays, end-point and quantitative reverse transcription PCR, and immunohistochemistry approaches.

Another variable aspect of studies predicting chemotherapy response is the computational and statistical approaches utilised. One of most popular methods for survival analysis is Cox proportional hazards regression. This model assumes that the hazard of death is proportional to the exponential of a linear predictor formed of the explanatory variables. This model has the advantage that, unlike many other regression techniques, it can appropriately deal with right-censored data such as that found in medical studies where patients leave before the end of the study period [15].

Other popular modelling techniques include linear models, support vector machines, hierarchical clustering, principal components analysis and the formation of a scoring algorithm. When dealing with data sets of varying sizes it is important to consider the number of samples and the amount of data per patient when choosing a modelling method. If the number of patients is large it is clear that a model will be better informed about the population from which the patient sample was drawn, and hence is likely to generalise more effectively to independent data sets. As the number of measurements per patient increases, the dimensionality and hence the flexibility of the model may increase. However, it is also important that the number of patients is sufficiently large to supply enough information about the factors being considered. Of the models identified here, linear models are relatively restrictive as the relationship between any factor and the outcome is assumed to be linear and so are suitable for smaller data sets. Conversely, hierarchical clustering simply finds groups of similar samples and there are minimal assumptions concerning the relationship between factors and outcome.

Classification models are used to predict which of a number of groups an individual falls into and are used for categorical variables, such as tumour grade and having or not having a disease. For visualisation and the assessment of classification model predictive power, a Kaplan-Meier plot is often combined with the log-rank test to investigate significance. It is worth noting that this method does not compare predictions with measurements, it simply considers the difference in survival between groups.

Many of the studies identified by this review involved developing a model using one set of samples, a training set, followed by testing of the model carried out on an independent set of samples, the test or validation set. This partitioning of samples is important as it allows the generalisability of the model to be assessed, and hence guards against over-fitting. If this check is not carried out, the true predictive ability of the model will not be known.

The aim of this review is to investigate the literature surrounding the prediction of chemotherapy response in ovarian cancer using gene expression. It has been observed, for example by Gillet et al. [16], that gene signatures obtained from cancer cell lines are not always relevant to in vivo studies, and that cell lines are inaccurate models of chemosensitivity [17]. The search was therefore restricted to studies involving human tissue in order to ensure that the resulting gene signatures are applicable in a clinical setting. It was also specified that the study must involve patients who have undergone chemotherapy treatment, so that the effects of resistance may be investigated.

Methods

Search methodology

The aim of this review is to investigate the literature on the prediction of chemoresistance in patients with ovarian cancer. Therefore, the six most important requirements identified were:

  • Concerned with (specifically) ovarian cancer

  • Patients were treated with chemotherapy

  • Gene expression was measured for use in predictions

  • Predictions are related to a measure of chemoresistance (e.g. response rates, progression-free survival)

  • Measurements were taken on human tissue (not cell lines)

  • The research aim is to develop a diagnostic tool or predict response

A PubMed search was carried out on 6th August 2014 to identify studies fulfilling the above requirements. The search terms may be found in Additional file 1. This search resulted in 78 papers.

Filtering

The search results were filtered twice, once based on abstracts and once based on full texts, by KL. An overview of the filtering process may be found in Figure 1. For the abstract-based filtering, papers were excluded if the six essential criteria were not all met, if the paper was a review article or if the paper was non-English language. This resulted in 48 papers remaining. For the full-text-based filtering, exclusion was due to not fulfilling the search criteria or papers that were not available. 42 papers were remaining after full-text-based filtering.

Figure 1.

Figure 1

PRISMA search filtering flow diagram. The initial search results were filtered using titles and abstracts and, later, the full text to ensure the search criteria were fulfilled. Following filtering the number of papers included reduced from 78 to 42.

Data extraction

Data was extracted using a pre-defined table created for the purpose. Extraction was carried out in duplicate by a single author (KL) with a wash-out period of 3 months to avoid bias. Variables extracted were: author, year, journal, number of samples, number of genes measured, study end-point, tissue source, percentage cancerous tissue, gene or protein expression measurement technique, sample histological types and stages, patient prior chemotherapy, modelling techniques applied, whether the model accounts for heterogeneity in patient chemotherapy, whether the model was prognostic or predictive, whether the model was validated, model predictive ability including any metrics or statistics, and the genes found to be predictive.

Bias analysis

Bias in the studies selected for the systematic review was assessed according to QUADAS-2 [18], a tool for the quality assessment of diagnostic accuracy studies. Levels of evidence were also assessed according to the CEBM 2011 Levels of Evidence [19]. Results of these analyses may be found in Additional files 2 and 3. Briefly, the majority of studies were considered to be low risk, with six studies judged to have unclear risk for at least one domain and seven studies judged to be high risk for at least one domain. Thirty-six studies where judged to have evidence of level 2, with the remaining six having evidence of level 3. These levels of risk and evidence suggest that the majority of conclusions drawn from these studies are representative and applicable to the review question.

Gene set enrichment

Gene set enrichment analysis was applied to the gene sets reported by the studies selected for this review. Analysis was performed using the R package HTSanalyseR [20]. Where reported, gene sets were extracted and combined according to the chemotherapy treatments applied to patients in each study. The two groups assessed were those studies where all patients were treated with platinum and taxane in combination, and those studies where patients were given treatments other than platinum and taxane. The second group includes those given platinum as a single agent. Any studies reporting treatments from both groups were excluded, as were studies that did not report the chemotherapy treatments used. Kyoto Encyclopedia of Genes and Genomes (KEGG) terms were identified for each gene and gene set collection analysis was carried out, which applies hypergeometric tests and gene set enrichment analysis. A p-value cut-off of 0.0001 was used. Enrichment maps were then plotted, using the 30 most significant KEGG terms. P-values were adjusted using the ‘BH’ correction [21].

Ethics statement

Ethical approval was not required for this systematic review, which deals exclusively with previously published data.

Results

Tables 1, 2, 3, 4, 5 and 6 detail some key information regarding the studies included in the review. Table 1 contains the number of samples analysed, the number of genes considered for the model, and the resulting genes retained as the predictive gene signature. Table 2 provides information about the tissue used for gene expression measurements and whether the studies assessed the percent neoplastic tissue before measurement, and Table 3 details the gene expression measurement techniques used. Table 4 contains the reported histological types and stages of the samples processed by each study. Table 5 provides information on chemotherapy treatments undergone by patients, whether the model was prognostic or predictive, and whether the model was validated using either an independent set of samples or cross validation. Table 6 lists the outcome to be predicted, the modelling techniques applied, and the predictive ability of the resulting model.

Table 1.

Journal and study information of papers included in the systematic review

Study Journal No. samples No. genes in study No. genes in signature
Jeong et al. [22] Anticancer Res. 487 612 388, 612
Lisowska et al. [23] Front. Oncol. 127 >47000 0
Roque et al. [24] Clin. Exp. Metastasis 48 1 1
Li et al. [3] Oncol. Rep. 44 1 1
Schwede et al. [25] PLoS ONE 663 2632 51
Verhaak et al. [26] J. Clin. Invest. 1368 11861 100
Obermayr et al. [27] Gynecol. Oncol. 255 29098 12
Han et al. [28] PLoS ONE 322 12042 349, 18
Hsu et al. [29] BMC Genomics 168 12042 134
Lui et al. [30] PLoS ONE 737 NS 227
Kang et al. [31] J. Nat. Cancer Inst. 558 151 23
Gillet et al. [32] Clin. Cancer Res. 80 356 11
Ferriss et al. [33] PLos ONE 341 NS 251, 125
Brun et al. [34] Oncol. Rep. 69 6 0
Skirnisdottir and Seidal [35] Oncol. Rep. 105 3 2
Brenne et al. [36] Hum. Pathol. 140 1 1
Sabatier et al. [37] Br. J. Cancer 401 NS 7
Gillet et al. [38] Mol. Pharmeceutics 32 350 18, 10, 6
Chao et al. [39] BMC Med. Genomics 6 8173 NS
Schlumbrecht et al. [40] Mod. Pathol. 83 7 2
Glaysher et al. [41] Br. J. Cancer 31 91 10, 4, 3, 5, 5, 11, 6, 6
Yan et al. [42] Cancer Res. 42 2 1
Yoshihara et al. [43] PLoS ONE 197 18176 88
Williams et al. [44] Cancer Res. 242 NS 15 to 95
Denkert et al. [45] J. Pathol 198 NS 300
Matsumura et al. [46] Mol. Cancer Res. 157 22215 250
Crijns et al. [47] PLoS Medicine 275 15909 86
Mendiola et al. [48] PLoS ONE 61 82 34
Gevaert et al. [49] BMC Cancer 69 ∼24000 ∼3000
Bachvarov et al. [50] Int. J. Oncol. 42 20174 155, 43
Netinatsunthorn et al. [51] BMC Cancer 99 1 1
De Smet et al. [52] Int. J. Gynecol. Cancer 20 21372 3000
Helleman et al. [53] Int. J. Cancer 96 NS 9
Spentzos et al. [54] J. Clin. Oncol. 60 NS 93
Jazaeri et al. [55] Clin. Cancer Res. 40 40033, 7585 85, 178
Raspollini et al. [56] Int. J. Gynecol. Cancer 52 2 2
Hartmann et al. [57] Clin. Cancer Res. 79 30721 14
Spentzos et al. [58] J. Clin. Oncol. 68 12625 115
Selvanayagam et al. [59] Cancer Genet. Cytogenet. 8 10692 NS
Iba et al. [60] Cancer Sci. 118 4 1
Kamazawa et al. [61] Gynecol. Oncol. 27 3 1
Vogt et al. [62] Acta Biochim. Pol. 17 3 0

If more than one value is given, the study used multiple different starting gene-sets or found multiple gene signatures. NS: Not Specified.

Table 2.

Tissue information of papers included in systematic review

Study Tissue source % Cancerous tissue
Jeong et al. [22]
Lisowska et al. [23] Fresh-frozen NS
Roque et al. [24] FFPE, Fresh-frozen min. 70%
Li et al. [3] FFPE NS
Schwede et al. [25]
Verhaak et al. [26]
Obermayr et al. [27] Fresh-frozen, Blood NS
Han et al. [28]
Hsu et al. [29]
Lui et al. [30]
Kang et al. [31]
Gillet et al. [32] Fresh-frozen min. 75%
Ferriss et al. [33] FFPE min. 70%
Brun et al. [34] FFPE NS
Skirnisdottir and Seidal [35] FFPE NS
Brenne et al. [36] Fresh-frozen effusion, Fresh-frozen min. 50%
Sabatier et al. [37] Fresh-frozen min. 60%
Gillet et al. [38] Fresh-frozen effusion NS
Chao et al. [39]
Schlumbrecht et al. [40] Fresh-frozen min. 70%
Glaysher et al. [41] FFPE, Fresh min. 80%
Yan et al. [42] Fresh-frozen NS
Yoshihara et al. [43] Fresh-frozen min. 80%
Williams et al. [44]
Denkert et al. [45] Fresh-frozen NS
Matsumura et al. [46] Fresh-frozen NS
Crijns et al. [47] Fresh-frozen median = 70%
Mendiola et al. [48] FFPE min. 80%
Gevaert et al. [49] Fresh-frozen NS
Bachvarov et al. [50] Fresh-frozen min. 70%
Netinatsunthorn et al. [51] FFPE NS
De Smet et al. [52] Not specified NS
Helleman et al. [53] Fresh-frozen median = 64%
Spentzos et al. [54] Fresh-frozen NS
Jazaeri et al. [55] FFPE, Fresh-frozen NS
Raspollini et al. [56] FFPE NS
Hartmann et al. [57] Fresh-frozen min. 70%
Spentzos et al. [58] Fresh-frozen NS
Selvanayagam et al. [59] Fresh-frozen min. 70%
Iba et al. [60] FFPE, Fresh-frozen NS
Kamazawa et al. [61] FFPE, Fresh-frozen NS
Vogt et al. [62] None specified NS

If more than one value is given, the study used tissue from multiple sources. NS: Not Specified.

Table 3.

Gene expression measurement techique information of papers included in systematic review

Study Immunohistochemistry TaqMan array q-RT-PCR Commercial microarray Custom microarray RT-PCR
Jeong et al. [22]
Lisowska et al. [23]
Roque et al. [24]
Li et al. [3]
Schwede et al. [25]
Verhaak et al. [26]
Obermayr et al. [27]
Han et al. [28]
Hsu et al. [29]
Lui et al. [30]
Kang et al. [31]
Gillet et al. [32]
Ferriss et al. [33]
Brun et al. [34]
Skirnisdottir and Seidal [35]
Brenne et al. [36]
Sabatier et al. [37]
Gillet et al. [38]
Chao et al. [39]
Schlumbrecht et al. [40]
Glaysher et al. [41]
Yan et al. [42]
Yoshihara et al. [43]
Williams et al. [44]
Denkert et al. [45]
Matsumura et al. [46]
Crijns et al. [47]
Mendiola et al. [48]
Gevaert et al. [49]
Bachvarov et al. [50]
Netinatsunthorn et al. [51]
De Smet et al. [52]
Helleman et al. [53]
Spentzos et al. [54]
Jazaeri et al. [55]
Raspollini et al. [56]
Hartmann et al. [57]
Spentzos et al. [58]
Selvanayagam et al. [59]
Iba et al. [60]
Kamazawa et al. [61]
Vogt et al. [62]

Table 4.

Histology information of papers included in systematic review

Study Sub-type Stage
Jeong et al. [22] Serous, Endometrioid, Adenocarcinoma I, II, III, IV
Lisowska et al. [23] Serous, Endometrioid, Clear cell, Undifferentiated II, III, IV
Roque et al. [24] Serous, Endometrioid, Clear cell, Undifferentiated, Mixed IIIC, IV
Li et al. [3] Serous, Endometrioid, Clear cell, Mucinous, Transitional II, III, IV
Schwede et al. [25] Serous, Endometrioid, Clear cell, Mucinous, Adenocarcinoma, OSE I, II, III, IV
Verhaak et al. [26] NS II, III, IV
Obermayr et al. [27] Serous, Non-serous II, III, IV
Han et al. [28] Serous, Endometrioid, Clear cell, Mucinous, Mixed, Poorly differentiated II, III, IV
Hsu et al. [29] NS III, IV
Lui et al. [30] Serous II, III, IV
Kang et al. [31] Serous I, II, III, IV
Gillet et al. [32] Serous III, IV
Ferriss et al. [33] Serous, Clear cell, Other III, IV
Brun et al. [34] Serous, Endometrioid, Clear cell, Mucinous, Other III, IV
Skirnisdottir and Seidal [35] Serous, Endometrioid, Clear cell, Mucinous, Anaplastic I, II
Brenne et al. [36] Serous, Endometrioid, Clear cell, Undifferentiated, Mixed II, III, IV
Sabatier et al. [37] Serous, Endometrioid, Clear cell, Mucinous, Undifferentiated, Mixed I, II, III, IV
Gillet et al. [38] Serous III, IV, NS
Chao et al. [39] NS NS
Schlumbrecht et al. [40] Serous III, IV
Glaysher et al. [41] Serous, Endometrioid, Clear cell, Mucinous, Mixed, Poorly differentiated IIIC, IV
Yan et al. [42] Serous, Endometrioid, Clear cell, Mucinous, Transitional II, III, IV
Yoshihara et al. [43] Serous III, IV
Williams et al. [44] Serous, Endometrioid, Undifferentiated III, IV
Denkert et al. [45] Serous, Non-serous, Undifferentiated I, II, III, IV
Matsumura et al. [46] Serous I, II, III, IV
Crijns et al. [47] Serous III, IV
Mendiola et al. [48] Serous, Non-serous III, IV
Gevaert et al. [49] Serous, Endometrioid, Mucinous, Mixed I, III, IV
Bachvarov et al. [50] Serous, Endometrioid, Clear cell II, III, IV
Netinatsunthorn et al. [51] Serous III, IV
De Smet et al. [52] Serous, Endometrioid, Mucinous, Mixed I, III, IV
Helleman et al. [53] Serous, Endometrioid, Clear cell, Mucinous, Mixed, Poorly differentiated I/II, III/IV
Spentzos et al. [54] Serous, Endometrioid, Clear cell, Mixed I, II, III, IV
Jazaeri et al. [55] Serous, Endometrioid, Clear cell, Mixed, Undifferentiated, Carcinoma II, III, IV
Raspollini et al. [56] Serous IIIC
Hartmann et al. [57] Serous, Endometrioid, Mixed II, III, IV
Spentzos et al. [58] Serous, Endometrioid, Clear cell, Mixed I, II, III, IV
Selvanayagam et al. [59] Serous, Endometrioid, Clear cell, Undifferentiated III, IV
Iba et al. [60] Serous, Endometrioid, Clear cell, Mixed I, II, III, IV
Kamazawa et al. [61] Serous, Endometrioid, Clear cell III, IV
Vogt et al. [62] NS NS

Entries in bold indicate that the study data set was comprised of at least 80% this type. NS: Not Specified.

Table 5.

Basic modelling and patient information of papers included in systematic review

Study Patient prior chemotherapy treatment Model accounts for the different chemotherapies? Prognostic or predictive? Model validated?
Jeong et al. [22] Platinum-based Predictive
Lisowska et al. [23] Platinum/Cyclophosphamide, Platinum/Taxane Prognostic
Roque et al. [24] NS Prognostic
Li et al. [3] Platinum/Cyclophosphamide, Platinum/Taxane Prognostic
Schwede et al. [25] NS Prognostic
Verhaak et al. [26] NS Prognostic
Obermayr et al. [27] Platinum-based Prognostic
Han et al. [28] Platinum/Paclitaxel Prognostic
Hsu et al. [29] Platinum/Paclitaxel
+ additional treatments Prognostic
Lui et al. [30] NS Prognostic
Kang et al. [31] Platinum/Taxane Prognostic
Gillet et al. [32] Carboplatin/Paclitaxel Prognostic
Ferriss et al. [33] Platinum-based Predictive
Brun et al. [34] NS Prognostic
Skirnisdottir and Seidal [35] Carboplatin/Paclitaxel Prognostic
Brenne et al. [36] NS Prognostic
Sabatier et al. [37] Platinum-based Prognostic
Gillet et al. [38] NS Prognostic
Chao et al. [39] NS Prognostic
Schlumbrecht et al. [40] Platinum/Taxane Prognostic
Glaysher et al. [41] Platinum, Platinum/Paclitaxel Predictive
Yan et al. [42] Platinum-based Prognostic
Yoshihara et al. [43] Platinum/Taxane Prognostic
Williams et al. [44] NS Predictive
Denkert et al. [45] Carboplatin/Paclitaxel Prognostic
Matsumura et al. [46] Platinum-based Predictive
Crijns et al. [47] Platinum, Platinum/
Cyclophosphamide, Platinum/Paclitaxel Prognostic
Mendiola et al. [48] Platinum/Taxane Prognostic
Gevaert et al. [49] NS Prognostic
Bachvarov et al. [50] Carboplatin/Paclitaxel,
Carboplatin/Cyclophosphamide, Cisplatin/Paclitaxel Prognostic
Netinatsunthorn et al. [51] Platinum/Cyclophosphamide Prognostic
De Smet et al. [52] Platinum/Cyclophosphamide, Platinum/Paclitaxel Prognostic
Helleman et al. [53] Platinum/Cyclophosphamide, Platinum-based Prognostic
Spentzos et al. [54] Platinum/Taxane Prognostic
Jazaeri et al. [55] Carboplatin/Paclitaxel, Cisplatin/Cyclophosphamide, Carboplatin/Docetaxel, Carboplatin Prognostic
Raspollini et al. [56] Cisplatin/Cyclophosphamide, Carboplatin/Cyclophosphamide, Carboplatin/Paclitaxel Prognostic
Hartmann et al. [57] Cisplatin/Paclitaxel, Carboplatin/Paclitaxel Prognostic
Spentzos et al. [58] Platinum/Taxane Prognostic
Selvanayagam et al. [59] Cisplatin/Cyclophosphamide, Carboplatin/Cyclophosphamide, Cisplatin/Paclitaxel Prognostic
Iba et al. [60] Carboplatin/Paclitaxel Prognostic
Kamazawa et al. [61] Carboplatin/Paclitaxel Prognostic
Vogt et al. [62] Etoposide, Paclitaxel/Epirubicin, Carboplatin/Paclitaxel Predictive

If more than one value is given, the study included patients treated with different treatments. NS: Not Specified.

Table 6.

Basic modelling information of papers included in systematic review

Study Prediction Prediction method Predictive ability
Jeong et al. [22] Overall Survival Student’s T test, Hierarchical clustering, Compound covariate predictor algorithm, Cox proportional hazards regression, Kaplan-Meier curves, Log-rank test, ROC analysis ‘Taxane-based treatment significantly affected OS for patients in the YA subgroup (3 year rate: 74.4% with taxane vs. 37.9% without taxane, p=0.005 by log-rank test)’, ‘estimated hazard ratio for death after taxane-based treatment in the YA subgroup was 0.5 (95% CI=0.31−−0.82,p=0.005)’
Lisowska et al. [23] Chemoresponse, Disease-Free Survival, Overall Survival Support vector machines, Kaplan-Meier curves, Log-rank test No genes found to be significant in the training set were significant in the test set, for chemoresponse, DFS or OS
Roque et al. [24] Overall Survival Kaplan-Meier curves, Log-rank test, Student’s T test ‘OS was predicted by increased class III β-tubulin staining by both tumor (HR3.66, 96%CI=1.11–12.1, p=0.03) and stroma (HR4.53, 95%CI=1.28–16.1, p=0.02)’
Li et al. [3] Chemoresponse (chemoresistant vs. chemosensitive) Correlation of p-CFL1 staining and chemoresponse ‘immunostaining of p-CFL1 was positive in 77.3% of chemosensitive and in 95.9% of the chemoresistant’ (p=0.014, U=157.5)
Schwede et al. [25] Stem cell-like subtype, Disease-Free Survival, Overall Survival ISIS unsupervised bipartitioning, Diagonal linear discriminant analysis, Gaussian mixture modelling, Kaplan-Meier curves, Log-rank test OS (p values): Dressman =0.0354, Crijns =0.021, Tothill =4.4E−7
Verhaak et al. [26] Poor Prognosis vs. Good Prognosis Significance analysis of microarrays, Single sample gene set enrichment analysis, Kaplan-Meier curves, Log-rank test Good or Poor prognosis, likelihood ratio =44.63
Obermayr et al. [27] Disease-Free Survival, Overall Survival Kaplan-Meier curves, Cox proportional hazards regression, χ2 test ‘The presence of CTCs six months after completion of the adjuvant chemotherapy indicated relapse within the following six months with 41% sensitivity, and relapse within the entire observation period with 22% sensitivity (85% specificity)’
Han et al. [28] Complete Response or Progressive Disease Supervised principal component method 349 gene signature: ROC AUC =0.702, p=0.022. 18 gene: ROC AUC =0.614, p=0.197.
Hsu et al. [29] Progression-Dree Survival Semi-supervised hierarchical clustering Good Response vs. Poor Response, p=0.021
Lui et al. [30] Chemosensitivity, Overall Survival, Progression-Dree Survival Predictive score using weighted voting algorithm, Kaplan-Meier curves, Log-rank Test, Cox proportional hazards regression Response of 26 of 35 patients in an independent data set was correctly predicted, patients in the low-scoring group exhibited poorer PFS (HR=0.43, p=0.04), ROC AUC = 0.90(0.86–0.95)
Kang et al. [31] Overall Survival, Progression-Free Survival, Recurrence-Free Survival Kaplan-Meier curves, Log-rank test, Cox proportional hazards regression, Pearson correlation coefficient Berchuck dataset: HR=0.33, 95%CI=0.13–0.86, p=0.013; Tothill dataset: HR=0.61, 95%CI=0.36–0.99, p=0.044
Gillet et al. [32] Overall Survival, Progression-Free Survival Supervised principle components method, Cox proportional hazards regression, Kaplan-Meier curves, Log-rank test ‘An 11-gene signature whose measured expression significantly improves the power of the covariates to predict poor survival’(p<0.003)
Ferriss et al. [33] Overall Survival COXEN coefficient, Mann-Whitney U test, ROC analysis, Unsupervised Hierarchical Clustering Carboplatin: sensitivity = 0.906, specificity = 0.174, PPV = 60%, NPV = 57% (UVA-55 validation set)
Brun et al. [34] 2-year Disease-Free Survival Student’s T test, Principal component analysis, Concordance index, Kaplen-Meier curves, Log-rank test No genes were found to have prognostic value
Skirnisdottir and Seidal [35] Recurrence, Disease-Free Survival χ2 test, Kaplan-Meier curves, Log-rank test, Logistic regression, Cox proportional hazards regression p53-status (OR=4.123, p=0.009; HR=2.447, p=0.019) was a significant and independent factor for tumor recurrence and DFS.
Brenne et al. [36] OC or MM, Progression-Free Survival, Overall Survival Mann-Whitney U test, Kaplan-Meier curves, Log-rank test, Cox proportional hazards regression Cox Multivariate Analysis: EHF mRNA expression in pre-chemotherapy effusions was an independent predictor of PFS (p=0.033, relative risk=4.528)
Sabatier et al. [37] Progression-Free Survival, Overall Survival Cox proportional hazards regression, Pearson’s coefficient correlation score Favourable vs. Unfavourable: ‘sensitivity = 61.6%, specificity = 62.4%, OR=2.7, 95%CI=1.7–4.2; p=6.1×10−06, Fisher’s exact test’
Gillet et al. [38] Overall Survival, Progression-Free Survival, Treatment Response Linear regression, Hierarchical clustering, Kaplan-Meier curves, Log-rank test ‘6 gene signature alone can effectively predict the progression-free survival of women with ovarian serous carcinoma (log-rank p=0.002)’
Chao et al. [39] Chemoresistance Interaction and expression networks for pathway identification, pathway intersections, betweenness and degree centrality, Student’s T test No statistical measure available. Many genes identified have previously been found experimentally
Schlumbrecht et al. [40] Overall Survival, Recurrence-Free Survival Linear regression, Logistic regression, Cox proportional hazards regression, Kaplan-Meier curves, Unsupervised cluster analysis, Log-rank test, Mann-Whitney U test, χ2 test ‘Greater EIG121 expression was associated with shorter time to recurrence (HR=1.13 (CI=1.02–1.26), p=0.021)’, ‘Increased expression of EIG121 demonstrated a statistically significant association with worse OS (HR=1.21 (CI1.09–1.35), p<0.001)’
Glaysher et al. [41] Chemosensitivity AIC gene selection, Multiple linear regression Cisplatin: Radj2=0.836, p<0.001
Yan et al. [42] Chemosensitivity ANOVA, Student’s T test, Mann-Whitney U test ‘Immunostaining scores [Annexin A3] are significantly higher in platinum-resistant tumors (p=0.035)’
Yoshihara et al. [43] Progression-Free Survival Cox proportional hazards regression, Ridge regression, Prognostic index, ROC analysis, Kaplan-Meier curves, Log-rank test ‘Prognostic index was an independent prognostic factor for PFS time (HR=1.64, p=0.0001)’, sensitivity = 64.4%, specificity = 69.2%
Williams et al. [44] Overall Survival COXEN score, Kaplan-Meier curves, Student’s T test, ROC analysis, Spearman’s rank correlation coefficient, Logistic regression, Log-rank test Carboplatin and Taxol: sensitivity = 77%, specificity = 56%, PPV=71%, NPV=78%
Denkert et al. [45] Overall Survival Semi-supervised analysis via Cox scoring, Principal components analysis, Kaplan-Meier curves, Log-rank test, Cox proportional hazards regression Duke et al.: ‘clinical outcome is significantly different depending on the OPI (p=0.021), with an HR of 1.7 (CI 1.1–2.6)’
Matsumura et al. [46] Taxane sensitivity, Overall Survival Hierarchical clustering, Kaplan-Meier curves, Log-rank test ‘Patients in the YY1-High cluster who were treated with paclitaxel showed improved survival compared with the other groups (p=0.010)’
Crijns et al. [47] Overall Survival Supervised principal components method, Cox proportional hazards regression, Kaplan-Meier curves, Log-rank test, χ2 test OSP: (High-risk vs. low-risk) HR=1.940, CI=1.190–3.163, p=0.008
Mendiola et al. [48] Progression-Free Survival, Overall Survival Kaplan-Meier curves, Log-rank test, AIC-based model selection, ROC curves, Cox proportional hazards regression OS: sensitivity = 87.2%, specificity = 86.4%
Gevaert et al. [49] Platin Resistance/Sensitivity, Stage Principal component analysis, Least squares support vector machines Platin-Resistance/Sensitivity: sensitivity = 67%, specificity = 40%, accuracy = 51.11%
Bachvarov et al. [50] Chemoresistance Hierarchical Clustering, Support vector machines No prediction metric applied
Netinatsunthorn et al. [51] Overall Survival, Recurrence-Free Survival Kaplan-Meier curves, Cox proportional hazards regression OS: HR=1.98, 95%CI=1.28–3.79, p=0.0138 ; RFS: HR=3.36, 95%CI=1.60–7.03, p=0.0017
De Smet et al. [52] Stage I vs. Advanced stage, Platin-sensistive vs. Platin-resistant Principal component analysis, Least squares support vector machines Estimated Classification Accuracy: Stage I vs Advanced Stage =100%, Platin-sensitive vs. Platin-resistant =76.9%
Helleman et al. [53] Chemoresponse (responder vs. non-responder) Class prediction, Hierarchical clustering, Principal component analysis Test set: PPV=24%, NPV=97%, sensitivity =89%, specificity =59%
Spentzos et al. [54] Chemoresponse (pathological-CR or PD), Disease-Free survival, Overall Survival Class prediction analysis, Compound covariate algorithm, Average linkage hierarchical clustering, Kaplan-Meier curves, Log-rank test, Cox proportional hazards regression Cox PH (resistant vs. sensitive): Recurrence HR=2.7 (95%CI=1.2–6.1), Death HR=3.9 (95%CI=3.1–11.4)
Jazaeri et al. [55] Clinical response Class prediction 9 most significantly differentially expressed genes, primary chemoresistant vs. primary chemosensitive: accuracy =77.8%
Raspollini et al. [56] Overall Survival (high vs. low) Univariate logistic regression, χ2 test COX-2: OR=0.23, 95%CI=0.06–0.77, p=0.017; MDR1: OR=0.01, 95%CI=0.002–0.09, p=<0.0005
Hartmann et al. [57] Time To Relapse (early vs.late) Support vector machine, Kaplan-Meier curves, Log-rank test, average linkage clustering Accuracy =86%, PPV=95%, NPV=67%
Spentzos et al. [58] Disease-Free Survival, Overall Survival Supervised pattern recognition/class prediction, Kaplan-Meier curves, Log-rank test, Cox proportional hazards regression Unfavourable vs. Favourable OS : (CPH) HR=4.6, 95%CI=2.0–10.7, p=0.0001
Selvanayagam et al. [59] Chemoresistance (chemoresistant vs. chemosensitive) Supervised voice-pattern recognition algorithm (clustering) PPV=1, NPV=1
Iba et al. [60] Chemoresponse, Overall Survival Kaplan-Meier curves, Log-rank test, Cox propotionate hazards regression, ROC analysis, χ2 test, Student’s T test, Mann-Whitney U test ‘Patients with c-myc expression of over 200 showed a significantly better 5-year survival rate (69.8% vs. 43.5%)’, p<0.05
Kamazawa et al. [61] Chemoresponse (CR or PR vs. NC or PD) Defined threshold expressionto divide responders and non-responders MDR-1 (all samples): specificity =95%, sensitivity = 100%, predictive value =96%
Vogt et al. [62] Chemoresistance Correlation of AUC from in-vitro ATP-CVA and gene expression All p values for correlation of drugs and genes were >0.05

If more than one value is given, the study used multiple different prediction methods or predicted more than one endpoint.

Tissue source

For studies involving RNA extraction the tissue source is an important consideration, as RNA degradation and fragmentation could affect the results of techniques involving amplification. This is a notable issue in formalin fixed paraffin embedded (FFPE) tissue, due to the cross-linking of genetic material and proteins [63]. Of the 42 papers included in this review, the majority used fresh-frozen biopsy tissue. The numbers of each tissue source may be found in Table 7, and the tissue source used by individual papers may be found in Table 2. Nine papers did not use an RNA source directly as secondary data was used. Data sources were mostly other studies or data repositories, such as the TCGA dataset. Two studies did not specify the source tissue though extraction and expression measurement methods were detailed.

Table 7.

Numbers of studies using various mRNA sources

mRNA source Number of studies
FFPE tissue 12
Fresh-frozen tissue 22
Fresh-frozen effusion 2
Fresh tissue 1
Blood 1
Not used 9
Not specified 2

The majority of papers in this review used fresh-frozen tissue. This choice was likely made to minimise RNA degradation and hence improve measurement accuracy. Due to the risk of RNA degradation because of long storage times and the fixing process applied to FFPE tissue, it is often expected that FFPE tissue will be irreversibly cross-linked and fragmented. However, following investigation into RNA integrity when extracted from paired FFPE and fresh-frozen tissue, Rentoft et al. [64] found that for most samples up- and down-regulation of four genes was found to be the same whether measured in FFPE or fresh-frozen tissue. They concluded that, if samples were screened to ensure RNA quality, FFPE material can successfully provide RNA for gene expression measurement.

The use of fresh-frozen tissue in a research setting is not unusual, as can be seen from the fact that this tissue type was most popular in this review. However, for translational research expected to lead to a clinical test, this is not as reasonable. FFPE tissue is much more readily available, due to simpler acquisition and storage, and tissue is already taken for histological analysis. Therefore a model capable of using data obtained from FFPE tissue is much more likely to be applicable in a clinical setting.

Another important consideration is the proportion of neoplastic cells in the sample. For each paper the reported proportion may be seen in Table 2. Of the 42 papers, 14 reported that the proportion of cancerous cells was measured. This was usually done using hematoxylin and eosin stained histologic slides. It is important for the gene expression measurement that the tissue used contains a high proportion of neoplastic cells, and hence it is important that this pre-analytical variable is controlled. Of the studies in this review, those reporting the percentage cancerous cells were evenly distributed between FFPE and fresh-frozen tissues.

Gene or protein expression quantification

Of the studies highlighted by this review, there were four main techniques applied for gene or protein expression measurement: Probe-target hybridization microarrays, quantitative PCR, reverse transcription end-point-PCR, and immunohistochemical staining. Of these methods only immunohistochemistry measures protein expression, via classification of the level of staining, and the other methods quantify gene expression via measurement of mRNA copy number.

Methods involving probe-target hybridization are available commercially, and 19 of the 42 studies utilised these. For example the Affymetrix®; Human U133A 2.0 GeneChip and the Agilent®; Whole Human Genome Oligo Microarray were both used by multiple studies. Additionally, 7 studies used custom-made probe-target hybridization arrays. Probe-target hybridisation arrays generally measure thousands of genes and hence can provide a wealth data per sample. TaqMan®; microfluidic arrays or quantitative-PCR were used by 16 studies. These techniques are typically used for smaller panels of genes. The TaqMan®; arrays for example may contain up to 384 genes per array. These methods are more targeted and hence the price per sample is usually lower.

Immunohistochemistry is a more labour-intensive technique, requiring staining for each gene considered, and hence was mostly only used by studies using small numbers of genes. This technique, which is semi-quantitative due to the scoring systems employed, also suffers from a lack of standardisation of procedures. Of the 11 papers using this technique, the maximum number of genes analysed was seven, and the mean number of genes assessed was 2.8. Although these studies provide useful information regarding the correlation of particular genes with outcome, the small numbers of genes is likely to result in an incomplete gene signature and low predictive power.

Several of the papers utilising quantifiable techniques used an alternative method or replicates to obtain a measure of the assay variability. Five papers involving commercial or custom microarrays also used reverse transcription PCR (RT-PCR) to measure the expression of a small number of genes for comparison and one study used samples run in duplicate to calculate the coefficient of variation. Of the studies using TaqMan microfluidic arrays, two used samples run in duplicate to obtain the coefficient of variation. However, even fewer papers reported a metric representing the level of variability found. Two studies reported a coefficient of variation; Glaysher et al. [41] reported CoV=2%=0.02 for TaqMan arrays and Hartmann et al. [57] reported CoV=0.2 for their custom microarray. Another two reported Spearman’s or Pearson’s r coefficients of correlation between microarray and RT-PCR results. Yoshihara et al. [43] gave Pearson r values ranging from 0.5 to 0.8, and Crijns et al. [47] gave Spearman’s r values between -0.6 and -0.9.

Histology

Table 4 details the histology (types and stages) of the patient samples used by each study. As may be seen, the majority of studies were heterogeneous with respect to the types of cancer included. However, 23 of the 42 studies used at least 80% serous samples, suggesting that the majority of information contributed to the gene signatures of these studies is related to the mechanisms and pathways in serous cancer. In the authors’ opinion it is important to identify the histologies of patient samples: although treatment is currently the same across types, response to chemotherapy has been found to vary [9,65,66]. It therefore may be advisable for future studies to include histological information when developing models predicting chemotherapy response.

Chemotherapy

Table 5 lists the chemotherapy treatments undergone by patients in each study. The 10 papers labelled NS did not specify the regimen applied, though the patients did have chemotherapy. These cohorts cannot therefore be assumed to be homogeneous with respect to patient chemotherapy treatment. All studies that specified the chemotherapy regimen undergone by patients noted at least one platinum-based treatment. Of these, 24 included patients treated with a platinum-taxane combination and 10 with a cyclophosphamide-platinum combination. It is important to note that 19 of the 42 papers stated the population was heterogeneous with regards to chemotherapy treatments and, of those that did, only 8 included patient treatment history as a feature of the study. The aims of the majority of the studies were to identify genes of which the expression may be used to predict survival time, or prognosis. As already noted, the presence of resistance to the chemotherapy agent administered will dramatically affect the survival of a patient. It is therefore reasonable to expect the gene signatures identified to include genes responsible for chemoresistance, which will depend on the mechanism of action of the drug. Using a heterogeneous cohort in terms of chemotherapy treatment may then be causing problems with the identification of a minimal predictive gene set.

End-point to be predicted

As may be expected, there was variation between the end-point chosen by studies for prediction. Popular end-points include overall survival, progression-free survival and response to chemotherapy. The endpoints considered by each study may be found in Table 6. Of these some are clinical endpoints, such as overall survival, others use non-clinical endpoints, such as response to chemotherapy, many of which are considered to be surrogates for overall survival. For cancer studies, overall survival is considered to be the most reliable and is the variable that is of most interest when considering the effect of an intervention.

Model development

Within this review, many different modelling techniques were used to identify an explanatory gene signature to predict patient outcome. The most popular was Cox proportional hazards regression, which was applied by 17 studies. This was closely followed by hierarchical clustering, which was used by 11 studies. All other methods were used by 8 or fewer studies. In total 24 different types of modelling techniques were applied, ranging from statistical tests such as Student’s T test and Mann-Whitney U test, to logistic regression, to ridge regression. Table 8 lists the modelling techniques identified and the number of studies that employed them. It is of interest that most of the techniques applied are forms of classification. These methods result in samples being assigned to groups, such as ‘good prognosis’ and ‘poor prognosis’. Whilst this may be useful in some settings, for a clinically-applicable tool a regression technique may be more appropriate as it will provide a value, such as a likelihood of relapse, rather than simply a class. Techniques in Table 8 capable of a numeric prediction include logistic and linear regression, Cox proportional hazards regression, and ridge regression.

Table 8.

Key modelling techniques applied by studies in the review

Technique Number of papers
Cox proportional hazards regression 17
Hierarchical clustering 11
Principal components analysis 8
Student’s T test 7
Scoring algorithm 6
Support Vector Machines 5
Correlation coefficients 5
Mann-Whitney U test 5
χ2 test 5
ROC analysis 5
Class prediction 4
Logistic regression 3
Linear regression 3
AIC gene selection 2
Concordance index 1
Pathway interaction networks 1
ANOVA 1
Expression threshold identified 1
Gene set enrichment analysis 1
Linear discriminant analysis 1
ISIS bipartitoning 1
Gaussian mixture modelling 1
Significance analysis of microarrays 1
Ridge regression 1

Jointly with the modelling methods identified above, 23 of the 42 studies implemented Kaplan-Meier curves to visualise the survival of the patient classes identified by the models. This enables the difference in survival between classes, for example ‘good prognosis’ and ‘poor prognosis’, to be seen and assessed. The application of a log-rank test assesses the separation of the curves and identifies whether there is a statistically significant difference in survival distribution between the classes. It should be noted that, although this gives an idea of separation of classes achieved by the model, the model results must still be compared with known outcomes to check positive and negative predictive power. This step was missing in several papers, such as Gillet et al. [38], where the p value returned by the log-rank test is given as the measure of model success.

It is important to highlight the difference between prognostic and predictive models. A prognostic model is one capable of predicting prognosis, such as survival time, using patient information and biomarkers and does not vary between different treatment options. In contrast, a predictive model is one able to predict the effect of a treatment on patient prognosis [67,68]. It is therefore clear that, although prognostic models may be useful for research purposes and when one treatment option is available (such as the standard platinum-taxane combination), predictive models have a much greater part to play in stratified medicine where the aim is to identify the most appropriate treatment on a patient-by-patient basis. In order for a model to be predictive, the effects of multiple treatments must be considered and the response compared with the biomarker status. Classification of the studies as prognostic or predictive may be seen in Table 5. Of the papers identified by this review, only a minority considered the effects of chemotherapy treatment on the predicted outcome and hence could be considered predictive. Glaysher et al. [41] and Vogt et al. [62] produced separate models for various treatments, allowing the effects of different drugs and combinations to be compared. Both studies applied drugs in vitro to cultured tissue to measure response to chemotherapy. This was combined with gene expression measurements to form the model training data set. In this way the same patient samples may be used to create a set of models predicting response to a variety of drugs. These models are therefore predictive rather than prognostic. Alternatively, models may be trained on sets of patients split by treatments undergone, which would lead to treatment-specific models predicting response to the particular drug. This method was used by Jeong et al. [22], Ferriss et al. [33], Williams et al. [44] and Matsumura et al. [46]. Additionally, the use of a model variable specifying patient treatment history could allow these models to be combined onto one using a single training set of all patients. The model may then be passed a variable specifying the drug of interest for resistance prediction. A simple version of this method was implemented by Crijns et al. [47], who included a feature for whether a patient was treated with paclitaxel. It is clear that the integration of patient chemotherapy treatment into these models is underused, and it is likely to be beneficial for this to be incorporated into future research.

Genes identified

Of the 42 papers in this review, 32 provided full or partial lists of the genes identified by their models. Of the remainder, it was common that the gene sets were large or that the genes were not explicitly identified by the model, as is the case with modelling techniques such as principal components analysis.

In total across the papers, 1298 unique genes were selected by models and of these 93.53% were found by only one paper. The most commonly chosen gene was selected by only four papers. Table 9 shows the numbers and percentages of genes chosen by one to four papers.

Table 9.

Numbers and percentages of genes featured in the gene sets of various numbers of papers

Number of papers Number of genes Percent of genes
identifying a gene
1 1214 93.53%
2 78 6.01%
3 5 0.385%
4 1 0.08%

A list of the genes identified by the papers in the review may be found in Table 10.

Table 10.

List of genes reported by studies included in this review

A1BG CHPF2 FSCN1 LRRC16B PKD1 SOBP
A2M CHRDL1 FXYD6 LRRC17 PKHD1 SORBS3
AADAC CHRNE FZD4 LRRC59 PLA2G7 SOS1
AAK1 CHST6 FZD5 LRSAM1 PLAA SOX12
ABCA13 CHTOP G0S2 LSAMP PLAU SOX21
ABCA4 CIAPIN1 G3BP1 LSM14A PLAUR SPANXD
ABCB1 CIB1 GABRP LSM3 PLCB3 SPATA13
ABCB10 CIB2 GAD1 LSM7 PLEC SPATA18
ABCB11 CIITA GALNT10 LSM8 PLEK SPATA4
ABCB7 CILP GAP43 LTA4H PLIN2 SPC25
ABCC3 CITED2 GART LTB PLS1 SPDEF
ABCC5 CKLF GATAD2A LTK PMM1 SPEN
ABCD2 CLCA1 GCH1 LUC7L2 PMP22 SPHK2
ABCG2 CLCNKB GCHFR LY6K PMVK SPOCK2
ABLIM1 CLDN10 GCM1 LY96 PNLDC1 SPTBN2
ACADVL CLIP1 GDF6 LZTFL1 PNLIPRP2 SRC
ACAT2 CNDP1 GFRA1 MAB21L2 PNMA5 SREBF2
ACKR2 CNKSR3 GGCT MAD2L2 POFUT2 SRF
ACKR3 CNN2 GGT1 MAGEE2 POLH SRRM1
ACO2 CNOT8 GJB1 MAGEF1 POLR3K SRSF3
ACOT13 CNTFR GLRX MAK POMP SSR1
ACP1 cofilin1 GMFB MAMLD1 POU2AF1 SSR2
ACRV1 COL10A1 GMPR MANF POU5F1 SSUH2
ACSM1 COL21A1 GNA11 MAP6D1 PPAP2B SSX2IP
ACSS3 COL3A1 GNAO1 MAPK1 PPAT ST6GALNAC1
ACTA2 COL4A4 GNAZ MAPK1IP1L PPCDC STC2
ACTB COL4A6 GNG4 MAPK3 PPCS STK38
ACTBL3 COL6A1 GNG7 MAPK8IP3 PPFIA3 STX12
ACTG2 COL7A1 GNL2 MAPK9 PPIC STX1B
ACTR3B COX8A GNMT MAPKAP1 PPIE STX7
ACTR6 CPD GNPDA1 MAPKAPK2 PPP1R1A STXBP2
ADAMDEC1 CPE GOLPH3 MARCKS PPP1R1B STXBP6
ADAMTS5 CPEB1 GPIHBP1 MARK4 PPP1R2 SUB1
ADIPOR2 CRCT1 GPM6B MATK PPP1R26 SULT1C2
ADK CREB5 GPR137 MB PPP2R3C SULT2B1
AEBP1 CRYAB GPT2 MBOAT7 PPP2R5C SUPT5H
AF050199 CRYBB1 GPX2 MCF2L PPP2R5D SUSD4
AF052172 CRYL1 GPX3 MCL1 PPP4R4 SUV420H1
AFM CRYM GPX8 MCM3 PPP6R1 SV2C
AFTPH CSE1L GRAMD1B MDC1 PRAP1 SYNM
AGFG1 CSPP1 GRB2 MDFI PRELP SYT1
AGR2 CSRP1 GRK6 MDK PRKAB1 SYT11
AGT CSRP3 GRM2 MDR-1 PRKCH SYT13
AIPL1 CST6 GRPEL1 MEA1 PRKCI TAC3
AKAP12 CST9L GRSF1 MEAF6 PRKD3 TAP1
AKR1A1 CT45A6 GSPT1 MECOM PROC TASP1
AKR1C1 CTA-246H3.1 GSTM2 MEF2B PROK1 TBCC
AKT1 CTNNBL1 GSTT1 MEGF11 PRPF31 TBP
AKT2 CTSD GTF2E1 MEST PRRX1 TCF15
ALCAM CUTA GTF2F2 METRN PRSS16 TCF7L2
ALDH5A1 CX3CL1 GTF2H5 METTL13 PRSS22 TENM3
ALDH9A1 CXCL1 GTPBP4 METTL4 PRSS3 TEX30
ALG5 CXCL10 GUCY1B3 MFAP2 PRSS36 TFF1
ALMS1 CXCL12 GYG1 MFSD7 PSAT1 TFF3
AMPD1 CXCL13 GYPC MGMT PSMB5 TFPI2
ANKHD1 CXCR4 GZMB MINOS1 PSMB9 TGFB1
ANKRD27 CYB5B GZMK MKRN1 PSMC4 THBS4
ANXA3 CYBRD1 H2AFX MLF2 PSMD1 TIAM1
ANXA4 CYP27A1 H3F3A MLH1 PSMD12 TIMM10B
AOC1 CYP2E1 HAP1 MLX PSMD14 TIMM17B
AP2A2 CYP3A7 HBG2 MMP1 PSME4 TIMP1
APC CYP4X1 HDAC1 MMP10 PTBP1 TIMP2
API5 CYP4Z1 HDAC2 MMP12 PTCH2 TIMP3
APOE CYP51A1 HECTD4 MMP13 PTEN TKTL1
AQP10 CYSTM1 HES1 MMP16 PTGDS TLE2
AQP5 CYTH3 HEY1 MMP17 PTGS2 TM9SF2
AQP6 D4S234E HHIPL2 MMP3 PTP4A1 TM9SF3
AQP9 DAP HIF1A MMP7 PTP4A2 TMCC1
ARAF DAPL1 HIP1R MMP9 PTPRN2 TMED5
ARAP1 DBI HIPK1 MPZL1 PTPRS TMEM139
AREG DCBLD2 HIST1H1C MRPL2 PWP2 TMEM14B
ARFGEF2 DCHS1 HK2 MRPL35 QPRT TMEM150A
ARHGAP29 DCK HLAA MRPL49 R3HDM2 TMEM161A
ARHGDIA DCTN5 HLADMB MRPS12 RAB26 TMEM259
ARL14 DCTPP1 HLADOB MRPS17 RAB27B TMEM260
ARL6IP4 DCUN1D4 HMBOX1 MRPS24 RAB40B TMEM45A
ARMC1 DCUN1D5 HMGCS1 MRPS9 RAB5B TMEM50A
ARNT2 DDB1 HMGCS2 MRS2 RAB5C TMPRSS3
ARPC4 DDB2 HMGN1 MSH2 RABIF TMSB15B
ASAP1 DDR1 HMOX2 MSL1 RAC1 TMTC1
ASAP3 DDX23 HNRNPA1 MSMO1 RAC3 TMX2
ASF1A DDX49 HNRNPUL2 MST1 RAD23A TNFRSF17
ASIP DEFB132 HOPX MT1G RAD51 TNS1
ASPA DERL1 HOXA5 MTCP1 RAD51AP1 TOMM40
ASPHD1 DFNB31 HOXB6 MTMR11 RANBP1 TONSL
ASS1 DHCR7 HPN MTMR2 RANGAP1 TOP1
ASUN DHRS11 HRASLS MTPAP RARRES2 TOP2A
ATM DHRS9 Hs.120332 MTUS1 RB1 TOX3
ATP1B3 DHX15 HS3ST1 MTX1 RBBP7 TP53
ATP5D DHX29 HS3ST5 MUS81 RBFA TP53TG5
ATP5F1 DIAPH3 HSD11B2 MUTYH RBM11 TP73
ATP5L DICER1 HSD17B11 MXD1 RBM39 TPD52
ATP6V0E1 DIRC1 HSPA1L MXI1 RCHY1 TPM2
ATP7B DKK1 HSPA4 MYBPC1 RER1 TPP2
ATP8A2 DLAT HSPA8 MYC RFC3 TPPP
AUP1 DLEU2 HSPB7 MYCBP RGL2 TPRKB
AURKA DLG1 HSPD1 MYL9 RGP1 TRA
AURKC DLG3 HTATIP2 MYO1D RGS19 TRAF3IP2
AVIL DLGAP4 HTN1 MYOM1 RHOT1 TRAM1
B3GALNT1 DLGAP5 HTR3A NANOS1 RHPN2 TRAPPC4
B3GNT2 DMRT3 ICAM1 NASP RIIAD1 TRAPPC9
B4GALT5 DNAH2 ICAM5 NBEA RIN1 TREML1
BAG3 DNAH7 ID1 NBL1 RIT1 TREML2
BAIAP2L1 DNAJB12 ID4 NBN RNF10 TRIAP1
BAK1 DNAJB5 IDI1 NCAM1 RNF13 TRIM27
BASP1 DNAJC16 IFIT1 NCAPD2 RNF14 TRIM49
BAX DNASE1L3 IGF1R NCAPG RNF148 TRIM58
BCHE DOCK3 IGFBP2 NCAPH RNF34 TRIML2
BCL2A1 DPH2 IGFBP5 NCKAP5 RNF6 TRIT1
BCL2L11 DPM1 IGHM NCOA1 RNF7 TRMT1L
BCL2L12 DPP7 IGKC NCOR2 RNF8 TRO
BCR-ABL DPYSL2 IGKV1-5 NCR2 RNGTT TRPV4
BEAN DRD4 IHH NCSTN RNPEPL1 TRPV6
BEST4 DTYMK IKZF4 NDRG2 ROBO1 TSPAN3
BFSP1 DUSP2 IL11RA NDST1 ROR1 TSPAN4
BFSP2 DUSP4 IL15 NDUFA12 ROR2 TSPAN6
BGN DUX3 IL17RB NDUFA9 RP13-347D8.3 TSPAN7
BHLHE40 DYNLT1 IL1B NDUFAB1 RP13-36C9.6 TSR1
BIN1 DYRK3 IL23A NDUFAF4 RPA3 TTC31
BIRC5 E2F2 IL27 NDUFB4 RPL23 TTLL6
BIRC6 ECH1 IL6 NDUFS5 RPL29P17 TTPAL
BLCAP EDF1 IL8 NEBL RPL31 TTYH1
BLMH EDN1 IMPA2 NETO2 RPL36 TUBB3
BMP8B EDNRA ING3 NEUROD2 RPP30 TUBB4A
BMPR1A EDNRB INHBA NFE2 RPS15 TUBB4Q
BNIP3 EEF1A2 INPP5A NFE2L3 RPS16 TUSC3
BOLA3 EFCAB14 INPP5B NFIB RPS19BP1 UBD
BPTF EFEMP2 INSR NFKBIB RPS24 UBE2I
BRCA1 EFNB2 INTS12 NFS1 RPS28 UBE2K
BRCA2 EGF INTS9 NID1 RPS4Y1 UBE2L3
BRSK1 EGFR IRF2BP1 NIT1 RPS6KA2 UBE4B
BTN3A3 EHD1 ISCA1 NKIRAS2 RPSA UBR5
BTNL9 EHF ISG20 NKX31 RRAGC UGT2B17
C11orf16 EI24 ITGAE NKX62 RRBP1 UGT8
C11orf74 EIF1 ITGB2 NLGN1 RRN3 UHRF1BP1
C12orf5 EIF2AK2 ITGB6 NOP5/58 RSL24D1 UMOD
C16orf89 EIF3K ITGB7 NOS3 RSU1 UPK1A
C17orf45 EIF4E2 ITLN1 NOTCH4 RTN4R UPK1B
C17orf53 EIF5 ITM2A NOV RXRB UQCRC2
C17orf70 ELF3 ITM2C NOX1 RYBP URI1
C1orf109 ELF5 ITPR2 NPAS3 RYR3 USP14
C1orf115 EML4 ITPRIP NPR1 S100A10 USP18
C1orf159 ENC1 JAG2 NPR3 S100A4 USP21
C1orf198 ENOPH1 JAK2 NPTX2 S100P UST
C1orf27 ENSA JAKMIP2 NPTXR SAMD4B UTP11L
C1orf68 ENTPD4 KCNB1 NPY SASH1 UTP20
C1QTNF3 EPB41L4A KCNE3 NRBP2 SCAMP3 UVRAG
C20orf199 EPCAM KCNH2 NRG4 SCARF1 VDR
C2orf72 EPHB2 KCNJ16 NRP1 SCG2 VEGFA
C4A EPHB3 KCNN1 NSFL1C SCGB1C1 VEGFB
C4BPA EPHB4 KCNN3 NSL1 SCGB3A1 VEZF1
C6orf120 EPOR KCTD1 NSMCE4A SCNM1 VPS39
C6orf124 ERBB3 KCTD5 NT5C3A SCO2 VPS52
C9orf3 ERCC8 KDELC1 NTAN1 SCUBE2 VPS72
C9orf47 ERMP1 KDELR1 NTF4 SDF2L1 VTCN1
CA13 ESF1 KDELR2 NUDT21 SEC14L2 VTI1B
CACNA1B ESM1 KDM4A NUDT9 SELT WBP2
CACNG6 ESR1 Ki67 NUS1 SEMA3A WBP4
CADM1 ESRP2 KIAA0125 OAS3 SENP3 WDR12
CALML3 ESYT1 KIAA0141 OASL SENP6 WDR45B
CAMK2B ETS1 KIAA0226 ODF4 SEPN1 WDR7
CAMK2N1 ETV1 KIAA0368 OGFOD3 SERPINB6 WDR77
CANX EVA1A KIAA1009 OGN SERPIND1 WIT1
CAP1 EXOC6B KIAA1033 OPA3 SERPINF1 WIZ
CAP2 EXTL1 KIAA1324 OR10A3 SERTAD4 WNK4
CAPN13 EYA2 KIAA1551 OR2AG1 SETBP1 WNT16
CAPN5 F2R KIAA2022 OR4C15 SF3A3 WT1
CASC3 FAAH KIAA4146 OR51B5 SF3B4 WTAP
CASP9 FABP1 KIF3A OR51I1 SGCB WWOX
CASS4 FABP7 KIFC3 OR6F1 SGCG XBP1
CATSPERD FADS1 KIT OR9G9 SGPP1 XPA
CC2D1A FADS2 KLF12 OSGEPL1 SH3PXD2A XPO4
CCBL1 FAM133A KLF5 OSGIN2 SHFM1 XYLT1
CCDC130 FAM135A KLHDC3 OSM SHOX Y09846
CCDC135 FAM155B KLHL7 OXTR SIDT1 YBX1
CCDC147 FAM174B KLK10 P2RX4 SIGLEC8 YIPF3
CCDC167 FAM19A4 KLK6 PABPC4 SIRT5 YIPF6
CCDC19 FAM211B KPNA3 PAGR1 SIRT6 YLPM1
CCDC53 FAM217B KPNA6 PAH SIVA1 YWHAE
CCDC9 FAM49B KRT10 PAK4 SIX2 YWHAZ
CCL13 FAM8A1 KRT12 PALB2 SKA3 ZBTB11
CCL2 FANCB KYNU PARD6B SLAMF7 ZBTB16
CCL28 FANCE L1TD1 PAX6 SLC12A2 ZBTB8A
CCM2L FANCF LAMB1 PBK SLC12A4 ZC3H13
CCNA2 FANCG LAMTOR5 PBX2 SLC14A1 ZCCHC8
CCNG2 FANCI LARP4 PBXIP1 SLC15A2 ZEB2
CCT6A FARP1 LAX1 PCF11 SLC1A1 ZFHX4
CCZ1 FAS LAYN PCGF3 SLC1A3 ZFP91
CD34 FASLG LBR PCK1 SLC22A5 ZFR2
CD38 FBXL18 LCMT2 PCNA SLC25A37 ZKSCAN7
CD44 FCGBP LCTL PCNXL2 SLC25A41 ZMYND11
CD46 FCGR3B LDB1 PCOLCE SLC25A5 ZNF106
CD70 FEN1 LDHB PCSK6 SLC26A9 ZNF12
CD97 FEZ1 LGALS4 PDCD2 SLC27A6 ZNF124
CDC42EP4 FGF2 LGR5 PDE3A SLC29A1 ZNF148
CDCA2 FGFBP1 LHB PDGFA SLC2A1 ZNF155
CDH12 FGFR1OP LHX1 PDGFRA SLC2A5 ZNF180
CDH19 FGFR1OP2 LIN28A PDGFRB SLC37A4 ZNF200
CDH3 FGFR2 LINGO1 PDP1 SLC39A2 ZNF292
CDH4 FHL2 LIPA PDSS1 SLC4A11 ZNF337
CDH5 FILIP1 LIPC PDZK1 SLC5A1 ZNF432
CDK17 FJX1 LIPG PEBP1 SLC5A3 ZNF467
CDK20 FKBP11 LMO3 PEX11A SLC5A5 ZNF48
CDK5R1 FKBP1B LMO4 PEX6 SLC6A3 ZNF503
CDK8 FKBP7 LOC100129250 PFAS SLC7A2 ZNF521
CDKN1A FLII LOC149018 PGAM1 SMAD2 ZNF569
CDY1 FLJ41501 LOC1720 PHF3 SMC4 ZNF644
CDYL2 FLNC LOC389677 PHGDH SMG1 ZNF71
CEACAM5 FLOT2 LOC642236 PHKA1 SMPD2 ZNF711
CEACAM6 FLT1 LOC646808 PHKA2 SNIP1 ZNF74
CEACAM7 FMN2 LOC90925 PI3 SNRPA1 ZNF76
CEP55 FMO1 LPAR6 PIC3CD SNRPC ZNF780B
CES1 FN1 LPCAT2 PIGC SNRPD3 ZYG11A
CES2 FOXA2 LPCAT4 PIGR SNX13
CFI FOXD4L2 LPHN2 PIK3CG SNX19
CH25H FOXJ1 LRIG1 PIP5K1B SNX7
CHIT1 FOXO3 LRIT1 PITRM1 SOAT2

Gene names have been standardised. Genes in bold were selected by more than two studies.

It is clear that the gene sets selected by the studies are very different and there is very little overlap. The genes chosen by two or more studies may be seen in Table 11. Many of these genes are known to have links to cancer, which may suggest that these genes are therefore implicated in ovarian cancer. It is possible that, although the genes selected varied, they in fact represent similar mechanisms. This could occur if there are large sets of highly covariate genes representing particular cellular processes and the genes in the signatures were simply random selections from these gene sets. The same gene being selected by multiple papers would then be unlikely, although the same information contribution would be made. It may then be more informative to assess and compare the mechanisms controlled by the genes chosen as part of the models.

Table 11.

Genes chosen most commonly by studies in review

Gene symbol Number of studies Function Expression links to cancer in literature
AGR2 4 Cell migration and growth Prostate, breast, ovarian, pancreatic
MUTYH 3 Oxidative DNA damage repair Colorectal
AKAP12 3 Subcellular compartmentation of PKA Colorectal, lung, prostate
TP53 3 Cell cycle regulation Breast
TOP2A 3 Required for DNA replication Breast, prostate, ovarian
FOXA2 3 Liver-specific transcription factor Lung, prostate
SRC 2 Regulation of cell growth Colon, liver, lung, breast, pancreatic
SIVA1 2 Pro-apoptotic protein Many cancers
ALDH9A1 2 Aldehyde dehydrogenase Many cancers
LGR5 2 Associated with stem cells Cancer stem cells
EHF 2 Epithelial differentiation and proliferation Prostate
BAX 2 Apoptotic activator Colon, breast, prostate, gastric, leukaemia
CES2 2 Intestine drug clearance Colorectal
CPE 2 Synthesis of hormones and neurotransmitters
FGFBP1 2 Cell proliferation, differentiation and migration Colorectal, pancreatic
TUBB4A 2 Component of microtubules
ZNF12 2 Transcription regulation
RBM39 2 Steroid hormone receptor-mediated transcription
RFC3 2 Required for DNA replication
GNPDA1 2 Triggers calcium oscillations in mammalian eggs
ANXA3 2 Regulation of cellular growth Prostate, ovarian
NFIB 2 Activates transcription and replication Breast
ACTR3B 2 Actin cyctoskeleton organisation Lung
YWHAE 2 Mediates signal transduction Lung, endometrial
CYP51A1 2 Drug metabolism and lipid synthesis
HMGCS1 2 Cholesterol synthesis and ketogenesis
ZMYND11 2 Transcriptional repressor
FADS2 2 Regulates unsaturation of fatty acids
SNX7 2 Family involved in intracellular trafficking
ARHGDIA 2 Regulates the GDP/GTP exchange reaction of the Rho proteins Prostate, lung,
NDST1 2 Inflammatory response Prostate, breast
AOC1 2 Catalyses degredation of such as histamine and spermidine
DAP 2 Positive mediator of programmed cell death
ERCC8 2 Transcription-coupled nucleotide excision repair
GUCY1B3 2 Catalyzes conversion of GTP to the second messenger cGMP
HDAC1 2 Control of cell proliferation and differentiation Prostate, breast, colorectal, gastric
HDAC2 2 Transcriptional regulation and cell cycle progression Cervical, gastric, colorectal
IGFBP5 2 Cell proliferation, differentiation, survival, and motility Breast
IL6 2 Transcriptional inflammatory response, B cell maturation Many cancers
LSAMP 2 Neuronal surface glycoprotein Osteosarcoma
MDK 2 Cell growth, migration, angiogenesis Many cancers
MYCBP 2 Stimulates the activation of E box-dependent transcription
S100A10 2 Transport of neurotransmitters Colorectal, lung, breast
SLC1A3 2 Glutamate transporter
NCOA1 2 Stimulates hormone-dependent transcription Breast, prostate
TIAM1 2 Modulates the activity of Rho GTP-binding proteins Many cancers
VEGFA 2 Angiogenesis, cell growth, cell migration, apoptosis Many cancers
RPL36 2 Component of ribosomal 60S subunit
LBR 2 Anchors lamina and heterochromatin to the nuclear membrane
ABCB1 2 ATP-dependent drug efflux pump for xenobiotic compounds Many cancers
FASLG 2 Required for triggering apoptosis in some cell types Many cancers
TIMP1 2 Extracellular matrix, proliferation, apoptosis Many cancers
FN1 2 Cell adhesion, motility, migration processes Many cancers
TGFB1 2 Proliferation, differentiation, adhesion, migration Prostate, breast, colon, lung, bladder
XPA 2 DNA excision repair Many cancers
ABCB10 2 Mitochondrial ATP-binding cassette transporter
POLH 2 Polymerase capable of replicating UV-damaged DNA for repair
ITGAE 2 Adhesion, intestinal intraepithelial lymphocyte activation
ZNF200 2 Zinc finger protein
COL3A1 2 Collagen type III, occurring in most soft connective tissues
ACKR3 2 G-protein coupled receptor
EPHB3 2 Mediates developmental processes Lung, colorectal
NBN 2 Double-strand DNA repair, cell cycle control
PCF11 2 May be involved in Pol II release following polymerisation
DFNB31 2 Sterocilia elongation, actin cystoskeletal assembly
BRCA2 2 Double-strand DNA repair Breast, ovarian
AADAC 2 Arylacetamide deacetylase
CD38 2 Glucose-induced insulin secretion Leukaemia
CHIT1 2 Involved in degradation of chitin-containing pathogens
CXCR4 2 Receptor specific for stromal-derived-factor-1 Breast, glioma, kidney, prostate
EFNB2 2 Mediates developmental processes
MECOM 2 Apoptosis, development, cell differentiation, proliferation Leukaemia
FILIP1 2 Controls neocortical cell migration Ovarian
HSPB7 2 Heat shock protein
LRIG1 2 Regulator of signaling by receptor tyrosine kinases Glioma
MMP1 2 Breakdown of extracellular matrix Gastric, breast
PSAT1 2 Phosphoserine aminotransferase
SDF2L1 2 Part of endoplasmic reticulum chaperone complex
TCF15 2 Regulation of patterning of the mesoderm
EPHB2 2 Contact-dependent bidirectional signaling between cells Colorectal
ETS1 2 Involved in stem cell development, cell senescence and death Many cancers
TRIM27 2 Male germ cell differentiation Ovarian, endometrial, prostate
MARK4 2 Mitosis, cell cycle control Glioma
B4GALT5 2 Biosynthesis of glycoconjugates and saccharides

Genes listed by number of papers selecting each gene. Gene function and links to cancer obtained via cursory literature search.

Gene set enrichment

The gene sets reported by the studies identified in this review were assessed to identify whether certain biological pathways and mechanisms featured more prominently according to the genes selected. Studies were split by chemotherapy treatments recieved by the patients, and the groups identified were platinum and taxane, and other treatments (such as platinum, cyclophosphamide and combinations). Studies that did not specify the chemotherapy treatments used were excluded. Studies falling into the platinum and taxane group were Han et al. [28], Kang et al. [31], Gillet et al. [32], Skirnisdottir and Seidal [35], Schlumbrecht et al. [40], Yoshihara et al. [43], Denkert et al. [45], Hartmann et al. [57], Iba et al. [60], and Kamazawa et al. [61]. Studies falling into the other treatments group were Obermayr et al. [27], Sabatier et al. [27], Yan et al. [42], Netinatsunthorn et al. [51], and Helleman et al. [53]. The results of the gene set enrichment using the KEGG system may be seen in Figures 2 and 3. From the plots, it may be seen that both groups identify several cancer-related pathways relevant to the drug mechanisms of action.

Figure 2.

Figure 2

Gene set enrichment networks for studies assessing ovarian cancer patients treated with platinum and taxane. Network maps of the 30 most enriched KEGG pathways. Node marker size signifies the number of genes in this category, and the thickness of edges indicate the Jaccard similarity coefficient between categories. Node markers are coloured according to adjusted p value as reported by the hypergeometric test, where darker red denotes more highly significant.

Figure 3.

Figure 3

Gene set enrichment networks for studies assessing ovarian cancer patients treated with treatments other than platinum and taxane. Network maps of the 30 most enriched KEGG pathways. Node marker size signifies the number of genes in this category, and the thickness of edges indicate the Jaccard similarity coefficient between categories. Node markers are coloured according to adjusted p value as reported by the hypergeometric test, where darker red denotes more highly significant.

It is informative to consider the KEGG terms in the context of the mechanisms of action of the chemotherapy drugs applied. Both groups contain patients treated with platinum single agents or platinum-containing combinations. It should therefore be expected that processes associated with the mechanism of action of platinum will be enriched. Once activated, the platinum binds to DNA and results in the formation of monoadducts, intra-strand crosslinking, inter-strand crosslinking and protein crosslinking. This DNA structure change affects the ability of the DNA to be unwound and replicated, resulting in the triggering of the G2-M DNA damage checkpoint and cell cycle arrest. The affected cell will attempt DNA repair and, if unsuccessful, undergo apoptosis [69]. Expected KEGG terms therefore include those relating to apoptosis and DNA damage.

From Figure 2, KEGG pathways highlighted for this group of studies include ten cancer-specific terms and six cancer-related terms. Here italics denote a KEGG term. The ErbB signalling pathway has been found to influence in proliferation, migration, differentiation and apoptosis in cancer [70] and overexpression of ERBB1 and ERBB2 have been implicated in head and neck and breast cancers. The neurotrophin signalling pathway is known to trigger MAPK and PI3K signalling, affecting differentiation, proliferation and development, and survival, growth, motility and angiogenesis respectively [71]. Altered expression of genes in this pathway has been found to correlate with poorer survival in colon, breast, lung and prostate cancers. Changes in expression of genes relating to focal adhesion, which is responsible for attachment of cells to the extracellular matrix, have been implicated in cancer migration, invasion, survival and growth [72]. The TGF-beta signalling pathway also regulates many cellular processes, including proliferation, cellular adhesion and motility, coregulation of telomerase function, regulation of apoptosis, angiogenesis, immunosuppression and DNA repair [73]. The p53 signalling pathway has many varied links to cancer. This pathway many be triggered by various stress signals and can result in several responses, including cell cycle arrest, apoptosis, the inhibition of angiogenesis and metastasis, and DNA repair [74]. Finally, nucleotide excision repair is known to promote cancer development when both up and down regulated. Down-regulation correlates is thought to increases susceptibility to mutation formation and hence the formation of cancer [75], whereas up-regulation has been found to correlate with resistance to platinum as the DNA damage caused by the chemotherapy agent is repaired [76].

The first group of studies considered patients treated with taxanes in addition to platinum. Taxanes act by stabilising tubulin, preventing the microtubule structure formation required for mitosis. This results in cell cycle arrest at the G2/M DNA damage checkpoint and apoptosis. Mechanisms for taxane resistance are, however, not well understood. Two suggested mechanisms include the increased expression of multidrug transporters, and changes in the expression of the β-tubulin isoforms [77]. Neither of these mechanisms seem to be enriched in the platinum and taxol group. In addition to the single-agent effects of platinum and taxanes, there is an additional synergistic effect [78]. However, this effect is also not well studied and hence the mechanisms by which this occurs are not clear.

The second group, as seen in Figure 3, was composed of studies applying chemotherapy treatments other than platinum and taxanes. This group is heterogeneous with respect to chemotherapy treatment, and mainly consists of studies reporting treatment as ‘platinum-based’. The other drug explicitly mentioned by studies in this group is cyclophosphamide. This drug is an alkylating agent and acts to form adducts in DNA [79]. This DNA damage triggers the G2/M DNA damage checkpoint, resulting in DNA repair or apoptosis. This suggests that the same DNA repair mechanisms related to platinum treatment are also relevant to cyclophosphamide. For this group, the KEGG pathway analysis shows that the gene set is enriched with 14 pathways related to cancer, in addition to two general cancer-related terms. The mTOR signalling pathway is downstream to the PI3K/AKT pathway and regulates growth, proliferation and survival [80]. The MAPK signalling pathway controls the cell cycle, and has been found to contribute to the control of proliferation, differentiation, apoptosis, migration and inflammation in cancer [81]. The chemokine signalling pathway has been found to regulate growth, survival and migration in addition to its role in inflammation [82]. Angiogenesis and vasculogenesis are known to be regulated by the VEGF signalling pathway [83], which is already the target of treatments such as bevacizumab. Purine metabolism is required for the production and recycling of adenine and guanine, and hence is required for DNA replication. This process is the target of chemotherapies such as methotrexate. The term drug metabolism – other enzymes is partially cancer related; this term refers to five drugs: azathioprine, 6-mercaptopurine, irinotecan, fluorouracil and isoniazid. Of these, two are chemotherapy treatments; irinotecan is a topoisomerase-I inhibitor and fluorouracil acts as a purine analogue. Also featuring in Figure 3 are apoptosis, ErbB signalling pathway, focal adhesion, neurotrophin signalling pathway, B cell receptor signalling pathway and Jak-STAT signalling pathway, all of which are known to be related to cancer.

Overall, the gene sets appear to be enriched for cancer-related resistance mechanisms [84]. However, when combined there is little evidence from this analysis to suggest that the signatures are capturing chemotherapy-specific mechanisms in addition to more general survival pathways. The DNA repair terms may suggest a response to platinum-based treatment, though the down-regulation of these mechanisms is also related to cancer development and resistance in general [85]. It is likely that, due to the varying reliability suggested by the bias analysis and the reported model development techniques, the signal-to-noise ratio of informative genes is low when the gene signatures are combined, preventing the identification of processes of interest.

Model predictive ability

Sensitivity and specificity

The comparison of the success of the various models is difficult, particularly due to the fact that many papers report different metrics as measures of model accuracy. Many of these are also incomplete, not providing enough information to fully describe the model. Ideally, models should be applied to an independent set of samples with known outcomes and performance measures on this data set reported. For classification models an informative set of measures would be positive predictive value, negative predictive value, specificity and sensitivity:

Sensitivity=ntrue positiventrue positive+nfalse negativeSpecificity=ntrue negativentrue negative+nfalse positivePPV=ntrue positiventrue positive+nfalse positiveNPV=ntrue negativentrue negative+nfalse negative

where ntrue positive is the number of true positive predictions, nfalse positive is the number of false positive predictions, ntrue negative is the number of true negative predictions and nfalse negative is the number of false negative predictions.

Together these provide information on true positive and negative rates as well as false positive and false negative rates, all of which are important when assessing the performance of a model.

Using the sensitivity and specificity the positive and negative likelihood ratios may be calculated and, using the prevalence of the condition in the test population, the probability of a patient having the condition based on the test results may be found, as in the equations below.

LR+ve=sensitivity1specificityLR-ve=1sensitivityspecificityP(Condition+|Test+)=P(Condition+)1P(Condition+)·LR+veP(Condition+)1P(Condition+)·LR+ve+1P(Condition+|Test)=P(Condition)1P(Condition)·LR-veP(Condition)1P(Condition)·LR-ve+1

These post-test probabilities are much easier to interpret and incorporate the prevalence of the condition. It should be noted that in order for the test to be applied in a clinical situation the pre-test probabilities used, P(Condition+) and P(Condition−), should be correct for the population of patients to whom the test will be applied. Here the sample prevalence from each study was used for convenience. However, it would be informative to recalculate P(Condition+|Test+) and P(Condition+|Test−) for the general population of ovarian cancer patients, as this would provide a better comparison between models.

Table 12 details the post-test probabilities of patients having a condition based on a positive or negative test result from the models developed by studies in this review. The papers appearing here are those that supplied sensitivity and specificity and the numbers of patients with and with without the condition, or alternative information allowing these to be calculated such as numbers of true and false positives and negatives.

Table 12.

Prediction metrics for studies reporting sensitivity and specificity

Study Prediction Sensitivity Specificity LR+ve LR-ve P(C+) P(C−) P(C+|T+) P(C+|T−)
Li et al. [3] Chemoresistance 0.96* 0.23* 1.24 0.18 2244 2244 0.55 0.15
Obermayr et al. [27] RFS 0.22* 0.85* 1.47 0.92 46216 170216 0.28 0.77
Ferriss et al. [33] Chemoresponse 0.94* 0.29* 1.33 0.20 85119 34119 0.77 0.07
Sabatier et al. [37] Prognosis 0.62* 0.62* 1.64 0.62 194366 172366 0.65 0.35
Yoshihara et al. [43] PFS 0.64* 0.69* 2.06 0.52 4587 3987 0.69 0.30
Williams et al. [44] Prognosis 0.77* 0.56* 1.75 0.41 97143 46143 0.79 0.16
Gevaert et al. [49] Chemoresistance 0.67* 0.40* 1.12 0.82 1545 3045 0.36 0.62
Helleman et al. [53] Chemoresistance 0.89* 0.56* 2.02 0.20 972 6372 0.22 0.58
De Smet et al. [52] Chemoresistance 0.71 0.83 4.29 0.34 613 713 0.79 0.29
Raspollini et al. [56] Prognosis 0.79 0.46 1.45 0.47 2852 2452 0.63 0.29
Hartmann et al. [57] Prognosis 0.86* 0.86* 6.14 0.16 2128 728 0.95 0.05
Selvanayagam et al. [59] Chemoresistance 1.00 1.00 0.00 48 48 1.00 0.00
Kamazawa et al. [61] Chemoresponse 1.00* 0.83 6.00 0.00 2127 527 0.95 0.00

*Value stated in reference.

Value calculated.

C: condition presence.

T: test result.

RFS: Relapse Free Survival.

PFS: Progression Free Survival.

From the table it may be seen that there is a great variety between the success of the models. For example, Kamazawa et al. [61] and Hartmann et al. [57] both achieved P(Condition+|Test+)=0.95 on their respective samples of the population. This means that if a patient tests positive, there is a 95% probability that they are positive for the condition in question, which in these cases are ‘responding to chemotherapy’ and ‘poor prognosis’ respectively. In contrast, Obermayr et al. [27], Helleman et al. [53] and Gevaert et al. [49] only achieved P(Condition+|Test+) of between 0.20 and 0.40. These results suggest that the tests are not able to predict the outcome of a patient any better than a random choice, and in the case of tests in the region of 0.20 it is likely that most patients are simply assigned to the same class.

The ability of tests to not commit type II errors and give false negatives is also important. Ferriss et al. [33] and Hartmann et al. [57] both achieved well in this regard, with P(Condition+|Test−)=0.07 and P(Condition+|Test−)=0.05 respectively. Several studies, by contrast, had very poor probabilities of false negatives; Obermayr et al. [27], Helleman et al. [53] and Gevaert et al. [49] all have P(Condition+|Test−)>0.5, which suggests that these models give a false negative more often than a random assignment.

Kamazawa et al. [61] and Selvanayagam et al. [59] both achieved extremely impressive prediction abilities, as may be seen by the very large P(Condition+|Test+) and very small P(Condition+|Test−) values. However, these studies exemplify why care must be taken in assessing the predictive ability of models. Both studies calculated sensitivity and specificity based on only training set results and hence there is no way to judge the generalisability of the models. There is a tendency for models to perform better on the training set than any following independent data set to which it is subsequently applied. Secondly, the training set used by Selvanayagam et al. [59] is extremely small at eight patients and has a 50 : 50 ratio of chemoresistant to chemosensitive patients. This sample is not representative of the population and hence the values of P(Condition+|Test+) and P(Condition+|Test−) will be skewed by unrepresentative P(Condition+) and P(Condition−).

Overall, the most successful model of this group is that by Hartmann et al. [57] as it makes predictions with good reliability and has been validated on an independent data set. The least successful models were Obermayr et al. [27], Helleman et al. [53] and Gevaert et al. [49]. These studies suffered from low ability to identify true positives and high probability of false positives, resulting in poor predictive ability.

Hazard ratios

It is common for studies of survival to quote hazard ratios comparing the results of clusters identified by classification models or relative-risk models such as Cox proportional hazards regression. These ratios represent the ratio of the probability of an event occurring to a patient in each of the two groups. The event is often death, but could also be recurrence for example. The studies listed in Table 13 supplied hazard ratios as measures of predictive ability. The hazard ratios vary from 0.23 to 4.6 with the majority around 2 to 3. A hazard ratio that is not equal to 1 suggests that the variable has predictive ability, and a ratio of 4, for example, suggests that a member of the high-risk group is 4 times as likely to die within the study period than a member of the low-risk group. The study with the highest hazard ratio is Spentzos et al. [58], with HR=4.6. This is closely followed by Raspollini [56] with HR=0.23 and Skirnisdottir and Seidal [35] with HR=4.12. The confidence intervals on the hazard ratios of all the studies are large and, with the exception of Spentzos et al. [58], at the lowest edge the hazard ratio is very close to 1. This suggests that, although all these hazard ratios were found to be significant, some were close to not reaching the arbitrary 5% level. Most notable are Roque et al. [24], Schlumbrecht and Seidal[40], and Denkert et al. [45]. These models would need further investigation to determine their predictive ability. Of the papers in this group, Spentzos et al. [58] appears to have the best predictive ability when classifying patients into two clusters with significantly different survival times.

Table 13.

Prediction metrics for studies reporting hazard ratios

Study Prediction Classes HR 95% CI Median survival P value
Jeong et al. [22] OS YA subgroup vs. YI subgroup 0.5 0.31−0.82 0.005
Roque et al. [24] OS High vs. low TUBB3 staining 3.66 1.11−12.05 707 days vs. not reached 0.03
Kang et al. [31] OS High vs. low score 0.33 0.13−0.86 1.8 years vs. 2.9 years <0.001
Skirnisdottir and Seidal [35] Recurrence p53 -ve vs. +ve 4.12 1.41−12.03 0.009
Schlumbrecht et al. [40] RFS EIG121 high vs. low 1.13 1.02−1.26 0.021
Yoshihara et al. [43] PFS High vs. low score 1.64 1.27−2.13 0.0001
Denkert et al. [45] OS Low vs. high score 1.7 1.1−2.6 0.021
Crijns et. al [47] OS 1.94 1.19−3.16 0.008
Netinatsunthorn et al. [51] RFS Yes vs. no WT1 staining 3.36 1.60−7.03 0.0017
Spentzos et al. [54] OS Resistant vs. sensitive 3.9 1.3−11.4 41 months vs. not reached <0.001
Raspollini et al. [56] OS No vs. yes COX-2 staining 0.23 0.06−0.77 0.017
Spentzos et al. [58] OS High vs. low score 4.6 2.0−10.7 30 months vs. not reached 0.0001

Calculated value.

HR: Hazard Ratio.

OS: Overall Survival.

RFS: Relapse Free Survival.

PFS: Progression Free Survival.

CI: Confidence Interval.

Linear regression

Two papers reported the success of model assessed using linear regression: Glaysher et al. [41] and Kang et al. [31]. These studies plotted the predicted values or model score against the measured values and applied linear regression to obtain a line of best fit. The R2 or Radj2 of this line is then calculated to assess the discrimination of the model. Glaysher et al. [41] achieved R2=0.901 (Radj2=0.836) for a model predicting resistance to cisplatin via cross-validation and Kang et al. [31] achieved R2=0.84 for a model predicting recurrence-free survival in the data set on which it was derived. These values suggest a good level of predictive ability, both in terms of calibration and discrimination, with the model by Glaysher et al. [41] achieving the better predictions.

Cox proportional hazards models

When studies identified by this review applied the Cox proportional hazards model to predict patient outcome, it was common for the main analysis of the model to be assessing whether the gene signature was found to be significant and whether the signature was an independent predictor. However, the application of this model to an independent data set was much less common. As may be seen from Table 6, the success of many models was judged using the significance of covariates including the gene signature in the model. It is likely that this model was not applied to external data sets due to subtleties in what the model predicts when compared to methods such as linear regression. Whereas in linear regression the survival times are predicted directly, Cox proportional hazards regression predicts hazard ratios. Royston and Altman [86] developed techniques for the external validation of Cox proportional hazards models by application to an independent data set. These rely on having at least the weights of the variables included in the linear predictor, and ideally the baseline survival function. The first allows the assessment of the discriminatory power of a model, whereas the second is also required to allow the calibration of the model to be assessed. Royston and Altman [86] are of the opinion that the inclusion of a log-rank test p-value is not informative due to the irrelevance of the null hypothesis being tested, and hence this should not be considered when judging model performance. An alternative to the log-rank test to compare survival between groups would be time-dependent ROC curves [87].

Failure to predict

Of the studies identified by this review, some models failed to achieve significant predictive ability. These include Lisowska et al. [23], Vogt et al. [62] and Brun et al. [34]. Of these papers, Vogt et al. [62] and Brun et al. [34] both considered small numbers of genes when constructing their models. It is possible then that these models failed because no informative genes were considered. Conversely, Lisowska [23] applied their modelling technique to over 47000 genes using 127 patients. It is therefore a possibility that genes were selected by their model purely by chance rather than due to true explanatory ability. This model was tested using an independent data. When the model was applied to this data set it performed poorly, suggesting that the genes chosen did not generalise to the second cohort of patients. Neither Vogt et al. [62] nor Brun et al. [34] reported measuring the precision or accuracy of the gene expression measurements. Lisowska et al. [23] used RT-PCR to measure the expression of 18 genes from the microarray, but the RT-PCR measurements were carried out on a separate set of samples and hence are not useful when considering accuracy. It is therefore unknown whether the gene expression measurement techniques applied by these studies were sufficiently accurate.

Discussion

The papers identified as part of this review tackled the important issue of chemoresistance and survival prediction in ovarian cancer via gene or protein expression. The concept of identifying gene signatures is popular, but requires careful handling to extract the information required for this to be successful. It was observed that of the many different tissue preservation techniques applied, the most common were fresh-frozen and formalin fixed, paraffin embedded tissue. It is our opinion that, due to the high quality expression measurements that may now be achieved with FFPE tissue, this is the most appropriate choice for research intended to translate into a clinical setting.

It was found that the majority of the studies included in this review were heterogeneous with respect to the histological type of the patient cohort. This suggests that, due to the differing response of different types of ovarian cancer to chemotherapy, the gene signatures may be identifying different pathways and mechanisms. However, it should also be noted that although 27 of the 42 studies were heterogeneous, 12 of these consisted of greater than 80% serous samples. Therefore, for these studies the inclusion of multiple histological types is likely to have less effect on the gene signature and mechanisms highlighted could be expected to occur in serous ovarian cancer. It would be advisable for future studies to include histological type and grade as model features.

The majority of studies identified by this review attempt to classify patients into groups with different characteristics, for example ‘poor prognosis’ and ‘good prognosis’ or ‘chemosensitive’ and ‘chemoresistant’. However, variables such as response to chemotherapy and prognosis are rarely so well separated into classes; they are by nature continuous variables. Altman and Royston [88] are clear that dichotomising continuous variables into categories (such as high-risk vs. low-risk) should be avoided, as it results in loss of information and may lead to underestimation of variation and the masking of non-linearity. Arbitrary choices of cutoff values may further obscure the situation, when the original continuous variable could serve the same purpose in many models. In terms of a clinical test it therefore may be more appropriate to apply alternative techniques, such as various types of regression, to obtain a real valued prediction of patient outcome.

It was noted that the metrics reported as measures of predictive ability vary between studies. These vary in the amount of information conveyed and hence care should be taken to use metrics that fully describe the model. Sensitivity and specificity are commonly reported for classification techniques and, together with the numbers of patients in each class in the data set, allows the probabilities of a patient having the condition of interest given that they have tested positive or negative. It is the ultimate aim of most classification studies to obtain these probabilities, as it allows the predictive ability of the test to be assessed and the applicability of the test to be evaluated. Of the studies reporting sensitivity, specificity and related information, the best predictive ability was achieved by Hartmann et al. [57] and the worst by Helleman et al. [53]. It is important to note that from the sensitivity and specificity the model by Helleman et al. [53] does not appear to be any worse than some of the others, but these probabilities incorporate the prevalence of the condition of interest in the test population. It would therefore be highly informative to recalculate these probabilities using the prevalence of the condition in the population of ovarian cancer patients. Since some of the test populations were not representative of the overall population (having so called ‘spectrum bias’), this would give a much more reliable indication of the predictive ability of the models in a clinical setting.

One of the main aims of the studies identified was to obtain a ‘gene signature’, the expression of which can explain and predict the response in the patient. To this end, the majority of the papers (32 of 42) provided full or partial list of the genes selected by the modelling process. An analysis of these gene signatures resulted in the conclusion that the signatures were very dissimilar, with the most commonly selected gene appearing in only four papers. 93.53% of genes were selected by only one paper. This seems to indicate that the gene signatures identified were not based on underlying cellular processes, or at least that the processes being highlighted were not the same across the papers. It should be noted that many of the studies used cohorts of patients who were heterogeneous in terms of chemotherapy treatment and, due to the development of resistance to chemotherapy via gene expression changes, this may affect the genes found to be explanatory. It may be that several gene signatures from sub-populations of patients treated with different drugs are combining and hence reducing the predictive ability of the models.

In order to assess the biological relevance of the genes selected for the gene signatures, gene set enrichment analysis was carried out. This technique is used to highlight processes and pathways that are over-represented in the gene signature compared to the set of all genes. For the purposes of this review, two groups of studies were considered: those where the patients were treated with platinum and taxane, and those where the patients were treated with other platinum based treatments. These groups were selected due to the low numbers of studies using a single treatment option. For example, there were no studies considering platinum, taxane or cyclophosphamide as single agents. Following the analysis, 30 KEGG terms were returned for each group. Of these, each list comprised of approximately half cancer related terms. Of these the majority were processes often up- or down-regulated in cancer cells, such as proliferation, apoptosis, and motility and metastasis [89]. It is unclear whether the change in regulation of these processes is further altered in response to specific chemotherapy treatments. However, one process worthy of additional consideration is DNA repair. DNA repair is known to be an important mechanism in cancer both though cancer development when down-regulated or mutated [75] and resistance to DNA damaging chemotherapy when up-regulated [76]. Therefore, the strong presence of DNA repair terms may suggest the presence of platinum resistance pathways in the gene signatures. It is the authors’ opinion that, although the combined gene signatures appear not to include predictive chemotherapy-specific information, they may be capable of providing prognostic information. It is also thought that some studies, such as Glaysher et al., may include genes relevant to additional chemotherapy-specific processes which are ‘drowned out’ when combined with other signatures.

Conclusion

It is clear that the prediction of response to chemotherapy in ovarian cancer is an ongoing research problem that has been attracting attention for many years. However, although many studies have been published, a clinical tool is still not available. It is our belief that, although not yet accomplished, progress within the field suggests that the development of a predictive model is possible. There is great variability between the approaches and success of existing studies in the literature, and there have been very high levels of variation in the genes identified as explanatory. It is the authors’ opinion that, if more care is taken when selecting the patients for inclusion to control for treatment history, these gene signatures may be simplified and models able to predict response to treatment may be developed.

Acknowledgements

KLL acknowledges support from an EPSRC PhD studentship (though MOAC DTC, EP/F500378/1). RSS acknowledges support from an MRC Biostatistics Fellowship.

Additional files

Additional file 1 (3.6KB, txt)

PubMed search terms.

Additional file 2 (114.8KB, pdf)

PRISMA Checklist.

Additional file 3 (70.4KB, pdf)

Bias assessment, including QUADAS-2 and CEBM levels of evidence.

Footnotes

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

IAC and RSS conceived and planned the study. Literature searches were carried out by KLL. KLL drafted the paper, which was critically reviewed and revised by IAC and RSS. All authors read and approved the final manuscript.

Contributor Information

Katherine L Lloyd, Email: K.Lloyd.1@warwick.ac.uk.

Ian A Cree, Email: I.A.Cree@warwick.ac.uk.

Richard S Savage, Email: R.S.Savage@warwick.ac.uk.

References

  • 1.Office for National Statistics: Cancer Incidence and Mortality in the United Kingdom. 2008–2010. http://www.ons.gov.uk/ons/rel/cancer-unit/cancer-incidence-and-mortality/2008-2010/rft-cancer-incidence-and-mortality-in-the-uk--2008-2010.xls.
  • 2.Office for National Statistics: Cancer Survival Rates, Cancer Survival in England, Patients Diagnosed 2005-2009 and Followed up to 2010. http://www.ons.gov.uk/ons/rel/cancer-unit/cancer-survival-rates/2005-2009--followed-up-to-2010/tbl-cancer-survival.xls.
  • 3.Li M, Yin J, Mao N, Pan L. Upregulation of phosphorylated cofilin 1 correlates with taxol resistance in human ovarian cancer in vitro and in vivo. Oncology Rep. 2013;29(1):58–66. doi: 10.3892/or.2012.2078. [DOI] [PubMed] [Google Scholar]
  • 4.Rubin SC, Randall TC, Armstrong KA, Chi DS, Hoskins WJ. Ten-year follow-up of ovarian cancer patients after second-look laparotomy with negative findings. Obstet Gynecol. 1999;93(1):21–4. doi: 10.1016/s0029-7844(98)00334-2. [DOI] [PubMed] [Google Scholar]
  • 5.Armstrong DK. Relapsed ovarian cancer: challenges and management strategies for a chronic disease. The Oncologist. 2002;7(Supplement 5):20–8. doi: 10.1634/theoncologist.7-suppl_5-20. [DOI] [PubMed] [Google Scholar]
  • 6.Di Nicolantonio F, Mercer SJ, Knight LA, Gabriel FG, Whitehouse PA, Sharma S, et al. Cancer cell adaptation to chemotherapy. BMC Cancer. 2005;5(1):78. doi: 10.1186/1471-2407-5-78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.National Institute for Health and Care Excellence: Guidance on the use of paclitaxel in the treatment of ovarian cancer. http://www.nice.org.uk/guidance/ta55.
  • 8.Goff BA. Advanced ovarian cancer: what should be the standard of care? J Gynecol Oncol. 2013;24(1):83–91. doi: 10.3802/jgo.2013.24.1.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Winter WE, Maxwell GL, Tian C, Carlson JW, Ozols RF, Rose PG, et al. Prognostic factors for stage iii epithelial ovarian cancer: a gynecologic oncology group study. J Clin Oncol. 2007;25(24):3621–7. doi: 10.1200/JCO.2006.10.2517. [DOI] [PubMed] [Google Scholar]
  • 10.Cancer Research UK: Ten-year survival over time. http://www.cancerresearchuk.org/prod_consump/groups/cr_common/@nre/@sta/documents/generalcontent/surv_10yrtrends_selcancers_xls.xls.
  • 11.Slodkowska EA, Ross JS. Mammaprint™ 70-gene signature: another milestone in personalized medical care for breast cancer patients. Expert Rev Mol Diagn. 2009;9(5):417–22. doi: 10.1586/erm.09.32. [DOI] [PubMed] [Google Scholar]
  • 12.Oncotype DX®;: Underlying Technology. http://breast-cancer.oncotypedx.com/en-US/Professional-Invasive/WhatIsTheOncotypeDXBreastCancerTest/UnderlyingTechnology.aspx.
  • 13.Oncotype DX®;: What Is the Colon cancer test?http://colon-cancer.oncotypedx.com/en-US/Professional/WhatIsTheColonCancerTest.aspx.
  • 14.Oncotype DX®;: Development. http://prostate-cancer.oncotypedx.com/en-US/Professional/IntroducingGPS/Development.aspx.
  • 15.Efron B. The efficiency of cox’s likelihood function for censored data. J Am Stat Assoc. 1977;72(359):557–65. doi: 10.1080/01621459.1977.10480613. [DOI] [Google Scholar]
  • 16.Gillet J-P, Calcagno AM, Varma S, Marino M, Green LJ, Vora MI, et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Nat Acad Sci. 2011;108(46):18708–13. doi: 10.1073/pnas.1111840108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Cree IA, Glaysher S, Harvey AL. Efficacy of anti-cancer agents in cell lines versus human primary tumour tissue. Curr Opin Pharmacol. 2010;10(4):375–9. doi: 10.1016/j.coph.2010.05.001. [DOI] [PubMed] [Google Scholar]
  • 18.Whiting PF, Rutjes AW, Westwood ME, Mallett S, Deeks JJ, Reitsma JB, et al. Quadas-2: a revised tool for the quality assessment of diagnostic accuracy studies. Ann Intern Med. 2011;155(8):529–36. doi: 10.7326/0003-4819-155-8-201110180-00009. [DOI] [PubMed] [Google Scholar]
  • 19.The Oxford 2011 Levels of Evidence (OCEBM Levels of Evidence Working Group). http://www.cebm.net/index.aspx?o=5653.
  • 20.Wang X, Terfve C, Rose JC, Markowetz F. Htsanalyzer: an r/bioconductor package for integrated network analysis of high-throughput screens. Bioinformatics. 2011;27(6):879. doi: 10.1093/bioinformatics/btr028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. Journal of the Royal Statistical Society. Series B (Methodological) 1995;57(1):289–300. [Google Scholar]
  • 22.Jeong W, Kim S-B, Sohn BH, Park Y-Y, PARK ES, Kim SC, et al. Activation of yap1 is associated with poor prognosis and response to taxanes in ovarian cancer. Anticancer Res. 2014;34(2):811–7. [PMC free article] [PubMed] [Google Scholar]
  • 23.Lisowska KM, Olbryt M, Dudaladava V, Pamula-Pilat J, Kujawa K, Grzybowska E, et al. Gene expression analysis in ovarian cancer - faults and hints from dna microarray study. Front Oncol. 2014;4:6. doi: 10.3389/fonc.2014.00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Roque DM, Buza N, Glasgow M, Bellone S, Bortolomai I, Gasparrini S, et al. Class iii beta-tubulin overexpression within the tumor microenvironment is a prognostic biomarker for poor overall survival in ovarian cancer patients treated with neoadjuvant carboplatin/paclitaxel. Clin Exp Metastasis. 2014;31(1):101–10. doi: 10.1007/s10585-013-9614-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Schwede M, Spentzos D, Bentink S, Hofmann O, Haibe-Kains B, Harrington D, et al. Stem cell-like gene expression in ovarian cancer predicts type ii subtype and prognosis. PLoS One. 2013;8:57799. doi: 10.1371/journal.pone.0057799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Verhaak RG, Tamayo P, Yang JY, Hubbard D, Zhang H, Creighton CJ, et al. Prognostically relevant gene signatures of high-grade serous ovarian carcinoma. J Clin Invest. 2013;123(1):517–25. doi: 10.1172/JCI65833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Obermayr E, Castillo-Tong DC, Pils D, Speiser P, Braicu I, Van Gorp T, et al. Molecular characterization of circulating tumor cells in patients with ovarian cancer improves their prognostic significance – a study of the ovcad consortium. Gynecol Oncol. 2013;128(1):15–21. doi: 10.1016/j.ygyno.2012.09.021. [DOI] [PubMed] [Google Scholar]
  • 28.Han Y, Huang H, Xiao Z, Zhang W, Cao Y, Qu L, et al. Integrated analysis of gene expression profiles associated with response of platinum/paclitaxel-based treatment in epithelial ovarian cancer. PLoS One. 2012;7:52745. doi: 10.1371/journal.pone.0052745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hsu FH, Serpedin E, Hsiao TH, Bishop AJ, Dougherty ER, Chen Y. Reducing confounding and suppression effects in tcga data: an integrated analysis of chemotherapy response in ovarian cancer. BMC Genomics. 2012;13 Suppl 6:13. doi: 10.1186/1471-2164-13-S6-S13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Liu Y, Sun Y, Broaddus R, Liu J, Sood AK, Shmulevich I, et al. Integrated analysis of gene expression and tumor nuclear image profiles associated with chemotherapy response in serous ovarian carcinoma. PLoS One. 2012;7:36383. doi: 10.1371/journal.pone.0036383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kang J, D’Andrea AD, Kozono D. A dna repair pathway-focused score for prediction of outcomes in ovarian cancer treated with platinum-based chemotherapy. J Natl Cancer Inst. 2012;104:670–81. doi: 10.1093/jnci/djs177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gillet JP, Calcagno AM, Varma S, Davidson B, Bunkholt Elstrand M, Ganapathi R, et al. Multidrug resistance-linked gene signature predicts overall survival of patients with primary ovarian serous carcinoma. Clin Cancer Res. 2012;18:3197–206. doi: 10.1158/1078-0432.CCR-12-0056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Ferriss JS, Kim Y, Duska L, Birrer M, Levine DA, Moskaluk C, et al. Multi-gene expression predictors of single drug responses to adjuvant chemotherapy in ovarian carcinoma: predicting platinum resistance. PLoS One. 2012;7:30550. doi: 10.1371/journal.pone.0030550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Brun JL, Cortez A, Lesieur B, Uzan S, Rouzier R, Darai E. Expression of mmp-2, -7, -9, mt1-mmp and timp-1 and -2 has no prognostic relevance in patients with advanced epithelial ovarian cancer. Oncol Rep. 2012;27(4):1049–57. doi: 10.3892/or.2011.1608. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Skirnisdottir I, Seidal T. The apoptosis regulators p53, bax and puma: Relationship and impact on outcome in early stage (figo i-ii) ovarian carcinoma after post-surgical taxane-based treatment. Oncol Rep. 2012;27(3):741–7. doi: 10.3892/or.2011.1578. [DOI] [PubMed] [Google Scholar]
  • 36.Brenne K, Nymoen DA, Hetland TE, Trope’ CG, Davidson B. Expression of the ets transcription factor ehf in serous ovarian carcinoma effusions is a marker of poor survival. Hum Pathol. 2012;43(4):496–505. doi: 10.1016/j.humpath.2011.05.023. [DOI] [PubMed] [Google Scholar]
  • 37.Sabatier R, Finetti P, Bonensea J, Jacquemier J, Adelaide J, Lambaudie E, et al. A seven-gene prognostic model for platinum-treated ovarian carcinomas. Br J Cancer. 2011;105:304–11. doi: 10.1038/bjc.2011.219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Gillet JP, Wang J, Calcagno AM, Green LJ, Varma S, Bunkholt Elstrand M, et al. Clinical relevance of multidrug resistance gene expression in ovarian serous carcinoma effusions. Mol Pharm. 2011;8(6):2080–8. doi: 10.1021/mp200240a. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Chao SY, Chiang JH, Huang AM, Chang WS. An integrative approach to identifying cancer chemoresistance-associated pathways. BMC Med Genomics. 2011;4:23. doi: 10.1186/1755-8794-4-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schlumbrecht MP, Xie SS, Shipley GL, Urbauer DL, Broaddus RR. Molecular clustering based on eralpha and eig121 predicts survival in high-grade serous carcinoma of the ovary/peritoneum. Mod Pathol. 2011;24:453–62. doi: 10.1038/modpathol.2010.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Glaysher S, Gabriel FG, Johnson P, Polak M, Knight LA, Parker K, et al. Molecular basis of chemosensitivity of platinum pre-treated ovarian cancer to chemotherapy. Br J Cancer. 2010;103:656–62. doi: 10.1038/sj.bjc.6605817. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yan X, Yin J, Yao H, Mao N, Yang Y, Pan L. Increased expression of annexin a3 is a mechanism of platinum resistance in ovarian cancer. Cancer Res. 2010;70:1616–24. doi: 10.1158/0008-5472.CAN-09-3215. [DOI] [PubMed] [Google Scholar]
  • 43.Yoshihara K, Tajima A, Yahata T, Kodama S, Fujiwara H, Suzuki M, et al. Gene expression profile for predicting survival in advanced-stage serous ovarian cancer across two independent datasets. PLoS One. 2010;5(3):9615. doi: 10.1371/journal.pone.0009615. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Williams PD, Cheon S, Havaleshko DM, Jeong H, Cheng F, Theodorescu D, et al. Concordant gene expression signatures predict clinical outcomes of cancer patients undergoing systemic therapy. Cancer Res. 2009;69:8302–9. doi: 10.1158/0008-5472.CAN-09-0798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Denkert C, Budczies J, Darb-Esfahani S, Gyorffy B, Sehouli J, Konsgen D, et al. A prognostic gene expression index in ovarian cancer - validation across different independent data sets. J Pathol. 2009;218(2):273–80. doi: 10.1002/path.2547. [DOI] [PubMed] [Google Scholar]
  • 46.Matsumura N, Huang Z, Baba T, Lee PS, Barnett JC, Mori S, et al. Yin yang 1 modulates taxane response in epithelial ovarian cancer. Mol Cancer Res. 2009;7:210–20. doi: 10.1158/1541-7786.MCR-08-0255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Crijns AP, Fehrmann RS, de Jong S, Gerbens F, Meersma GJ, Klip HG, et al. Survival-related profile, pathways, and transcription factors in ovarian cancer. PLoS Med. 2009;6:24. doi: 10.1371/journal.pmed.1000024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mendiola M, Barriuso J, Redondo A, Marino-Enriquez A, Madero R, Espinosa E, et al. Angiogenesis-related gene expression profile with independent prognostic value in advanced ovarian carcinoma. PLoS One. 2008;3(12):4051. doi: 10.1371/journal.pone.0004051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gevaert O, De Smet F, Van Gorp T, Pochet N, Engelen K, Amant F, et al. Expression profiling to predict the clinical behaviour of ovarian cancer fails independent evaluation. BMC Cancer. 2008;8:18. doi: 10.1186/1471-2407-8-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Bachvarov D, L’Esperance S, Popa I, Bachvarova M, Plante M, Tetu B. Gene expression patterns of chemoresistant and chemosensitive serous epithelial ovarian tumors with possible predictive value in response to initial chemotherapy. Int J Oncol. 2006;29(4):919–33. [PubMed] [Google Scholar]
  • 51.Netinatsunthorn W, Hanprasertpong J, Dechsukhum C, Leetanaporn R, Geater A. Wt1 gene expression as a prognostic marker in advanced serous epithelial ovarian carcinoma: an immunohistochemical study. BMC Cancer. 2006;6:90. doi: 10.1186/1471-2407-6-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.De Smet F, Pochet NL, Engelen K, Van Gorp T, Van Hummelen P, Marchal K, et al. Predicting the clinical behavior of ovarian cancer from gene expression profiles. Int J Gynecol Cancer. 2006;16 Suppl 1:147–51. doi: 10.1111/j.1525-1438.2006.00321.x. [DOI] [PubMed] [Google Scholar]
  • 53.Helleman J, Jansen MP, Span PN, van Staveren IL, Massuger LF, Meijer-van Gelder ME, et al. Molecular profiling of platinum resistant ovarian cancer. Int J Cancer. 2006;118(8):1963–71. doi: 10.1002/ijc.21599. [DOI] [PubMed] [Google Scholar]
  • 54.Spentzos D, Levine DA, Kolia S, Otu H, Boyd J, Libermann TA, et al. Unique gene expression profile based on pathologic response in epithelial ovarian cancer. J Clin Oncol. 2005;23:7911–8. doi: 10.1200/JCO.2005.02.9363. [DOI] [PubMed] [Google Scholar]
  • 55.Jazaeri AA, Awtrey CS, Chandramouli GV, Chuang YE, Khan J, Sotiriou C, et al. Gene expression profiles associated with response to chemotherapy in epithelial ovarian cancers. Clin Cancer Res. 2005;11:6300–10. doi: 10.1158/1078-0432.CCR-04-2682. [DOI] [PubMed] [Google Scholar]
  • 56.Raspollini MR, Amunni G, Villanucci A, Boddi V, Taddei GL. Increased cyclooxygenase-2 (cox-2) and p-glycoprotein-170 (mdr1) expression is associated with chemotherapy resistance and poor prognosis. analysis in ovarian carcinoma patients with low and high survival. Int J Gynecol Cancer. 2005;15:255–60. doi: 10.1111/j.1525-1438.2005.15212.x. [DOI] [PubMed] [Google Scholar]
  • 57.Hartmann LC, Lu KH, Linette GP, Cliby WA, Kalli KR, Gershenson D, et al. Gene expression profiles predict early relapse in ovarian cancer after platinum-paclitaxel chemotherapy. Clin Cancer Res. 2005;11:2149–55. doi: 10.1158/1078-0432.CCR-04-1673. [DOI] [PubMed] [Google Scholar]
  • 58.Spentzos D, Levine DA, Ramoni MF, Joseph M, Gu X, Boyd J, et al. Gene expression signature with independent prognostic significance in epithelial ovarian cancer. J Clin Oncol. 2004;22:4700–10. doi: 10.1200/JCO.2004.04.070. [DOI] [PubMed] [Google Scholar]
  • 59.Selvanayagam ZE, Cheung TH, Wei N, Vittal R, Lo KW, Yeo W, et al. Prediction of chemotherapeutic response in ovarian cancer with dna microarray expression profiling. Cancer Genet Cytogenet. 2004;154:63–6. doi: 10.1016/j.cancergencyto.2004.01.024. [DOI] [PubMed] [Google Scholar]
  • 60.Iba T, Kigawa J, Kanamori Y, Itamochi H, Oishi T, Simada M, et al. Expression of the c-myc gene as a predictor of chemotherapy response and a prognostic factor in patients with ovarian cancer. Cancer Sci. 2004;95(5):418–23. doi: 10.1111/j.1349-7006.2004.tb03225.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kamazawa S, Kigawa J, Kanamori Y, Itamochi H, Sato S, Iba T, 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: 10.1006/gyno.2002.6738. [DOI] [PubMed] [Google Scholar]
  • 62.Vogt U, Falkiewicz B, Bielawski K, Bosse U, Schlotter CM. Relationship of c-myc and erbb oncogene family gene aberrations and other selected factors to ex vivo chemosensitivity of ovarian cancer in the modified atp-chemosensitivity assay. Acta Biochim Pol. 2000;47(1):157–64. [PubMed] [Google Scholar]
  • 63.Medeiros F, Rigl CT, Anderson GG, Becker SH, Halling KC. Tissue handling for genome-wide expression analysis: a review of the issues, evidence, and opportunities. Arch Pathol Lab Med. 2007;131(12):1805–16. doi: 10.5858/2007-131-1805-THFGEA. [DOI] [PubMed] [Google Scholar]
  • 64.Rentoft M, Coates PJ, Laurell G, Nylander K. Transcriptional profiling of formalin fixed paraffin embedded tissue: pitfalls and recommendations for identifying biologically relevant changes. PloS One. 2012;7(4):35276. doi: 10.1371/journal.pone.0035276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Takano M, Tsuda H, Sugiyama T. Clear cell carcinoma of the ovary: is there a role of histology-specific treatment. J Exp Clin Cancer Res. 2012;31(53):35. doi: 10.1186/1756-9966-31-53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hess V, A’Hern R, Nasiri N, King DM, Blake PR, Barton DP, et al. Mucinous epithelial ovarian cancer: a separate entity requiring specific treatment. J Clin Oncol. 2004;22(6):1040–4. doi: 10.1200/JCO.2004.08.078. [DOI] [PubMed] [Google Scholar]
  • 67.Oldenhuis C, Oosting S, Gietema J, De Vries E. Prognostic versus predictive value of biomarkers in oncology. Eur J Cancer. 2008;44(7):946–53. doi: 10.1016/j.ejca.2008.03.006. [DOI] [PubMed] [Google Scholar]
  • 68.Tezak Z, Kondratovich MV, Mansfield E. Us fda and personalized medicine: in vitro diagnostic regulatory perspective. Personalized Med. 2010;7(5):517–30. doi: 10.2217/pme.10.53. [DOI] [PubMed] [Google Scholar]
  • 69.Tapia G, Diaz-Padilla I. Molecular mechanisms of platinum resistance in ovarian cancer In: Diaz-Padilla I, editor. Ovarian Cancer - A Clinical and Translational Update. InTech: 2013.
  • 70.Citri A, Yarden Y. Egf–erbb signalling: towards the systems level. Nat Rev Mol Cell Biol. 2006;7(7):505–16. doi: 10.1038/nrm1962. [DOI] [PubMed] [Google Scholar]
  • 71.Tan F, Thiele CJ, Zhijie L. Neurotrophin signaling in cancer. In: Kostrzewa RM, editor. Handbook of Neurotoxicity. New York: Springer; 2014. [Google Scholar]
  • 72.McLean GW, Carragher NO, Avizienyte E, Evans J, Brunton VG, Frame MC. The role of focal-adhesion kinase in cancer–a new therapeutic opportunity. Nat Rev Cancer. 2005;5(7):505–15. doi: 10.1038/nrc1647. [DOI] [PubMed] [Google Scholar]
  • 73.Elliott RL, Blobe GC. Role of transforming growth factor beta in human cancer. J Clin Oncol. 2005;23(9):2078–93. doi: 10.1200/JCO.2005.02.047. [DOI] [PubMed] [Google Scholar]
  • 74.Levine AJ, Bargonetti J, Bond GL, Hoh J, Onel K, Overholtzer M, et al. The p53 network. In: Zambetti GP, et al., editors. The P53 Tumor Suppressor Pathway and Cancer. US: Springer; 2005. [Google Scholar]
  • 75.Neumann AS, Sturgis EM, Wei Q. Nucleotide excision repair as a marker for susceptibility to tobacco-related cancers: A review of molecular epidemiological studies. Mol Carcinog. 2005;42(2):65–92. doi: 10.1002/mc.20069. [DOI] [PubMed] [Google Scholar]
  • 76.Martin LP, Hamilton TC, Schilder RJ. Platinum resistance: the role of dna repair pathways. Clin Cancer Res. 2008;14(5):1291–5. doi: 10.1158/1078-0432.CCR-07-2238. [DOI] [PubMed] [Google Scholar]
  • 77.Galletti E, Magnani M, Renzulli ML, Botta M. Paclitaxel and docetaxel resistance: molecular mechanisms and development of new generation taxanes. ChemMedChem. 2007;2(7):920–42. doi: 10.1002/cmdc.200600308. [DOI] [PubMed] [Google Scholar]
  • 78.Jekunen A, Christen R, Shalinsky D, Howell S. Synergistic interaction between cisplatin and taxol in human ovarian carcinoma cells in vitro. British J Cancer. 1994;69(2):299. doi: 10.1038/bjc.1994.55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hall A, Tilby M. Mechanisms of action of, and modes of resistance to, alkylating agents used in the treatment of haematological malignancies. Blood Rev. 1992;6(3):163–73. doi: 10.1016/0268-960X(92)90028-O. [DOI] [PubMed] [Google Scholar]
  • 80.Pópulo H, Lopes JM, Soares P. The mtor signalling pathway in human cancer. Int J Mol Sci. 2012;13(2):1886–918. doi: 10.3390/ijms13021886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Dhillon A, Hagan S, Rath O, Kolch W. Map kinase signalling pathways in cancer. Oncogene. 2007;26(22):3279–90. doi: 10.1038/sj.onc.1210421. [DOI] [PubMed] [Google Scholar]
  • 82.Hembruff SL, Cheng N. Chemokine signaling in cancer: Implications on the tumor microenvironment and therapeutic targeting. Cancer Ther. 2009;7(A):254. [PMC free article] [PubMed] [Google Scholar]
  • 83.Kowanetz M, Ferrara N. Vascular endothelial growth factor signaling pathways: therapeutic perspective. Clinical Cancer Res. 2006;12(17):5018–22. doi: 10.1158/1078-0432.CCR-06-1520. [DOI] [PubMed] [Google Scholar]
  • 84.Cree IA. Designing personalised cancer treatments. J Control Release. 2013;172(2):405–9. doi: 10.1016/j.jconrel.2013.07.007. [DOI] [PubMed] [Google Scholar]
  • 85.Bernstein C, Prasad AR, Nfonsam V, Bernstein H. Dna damage, dna repair and cancer In: Chen C, editor. New Research Directions in DNA Repair. InTech: 2013.
  • 86.Royston P, Altman DG. External validation of a cox prognostic model: principles and methods. BMC Med Res Methodol. 2013;13(1):33. doi: 10.1186/1471-2288-13-33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Heagerty PJ, Lumley T, Pepe MS. Time-dependent roc curves for censored survival data and a diagnostic marker. Biometrics. 2000;56(2):337–44. doi: 10.1111/j.0006-341X.2000.00337.x. [DOI] [PubMed] [Google Scholar]
  • 88.Altman DG, Royston P. Statistics notes: the cost of dichotomising continuous variables. BMJ: British Med J. 2006;332(7549):1080. doi: 10.1136/bmj.332.7549.1080. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100(1):57–70. doi: 10.1016/S0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]

Articles from BMC Cancer are provided here courtesy of BMC

RESOURCES