Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Aug 26.
Published in final edited form as: Cancer Biomark. 2010;8(0):231–251. doi: 10.3233/CBM-2011-0212

Prognostic Biomarkers in Ovarian Cancer

Jie Huang 1, Wei Hu 1, Anil K Sood 1,2,3,4
PMCID: PMC4143980  NIHMSID: NIHMS616102  PMID: 22045356

Abstract

Epithelial ovarian cancer (EOC) remains the most lethal gynecological malignancy despite several decades of progress in diagnosis and treatment. Taking advantage of the robust development of discovery and utility of prognostic biomarkers, clinicians and researchers are developing personalized and targeted treatment strategies. This review encompasses recently discovered biomarkers of ovarian cancer, the utility of published prognostic biomarkers for EOC (especially biomarkers related to angiogenesis and key signaling pathways), and their integration into clinical practice.

Keywords: biomarkers, ovarian carcinoma, prognosis

1. Introduction

Ovarian cancer remains the most lethal gynecologic cancer due, in part, to the advanced stage at presentation in most patients.1 For these patients, surgery followed by chemotherapy remains the standard of care.2 Given the diversity in responses, there is a need for highly reliable predictive and prognostic biomarkers. Since the introduction of CA125 as a biomarker for epithelial ovarian cancer (EOC) in 1981, the field of tumor biomarkers has grown substantially. Prognostic markers can help to identify patients at different levels of risk for specific outcomes, facilitate treatment choice, and aid patient counseling.3 In oncology, hundreds of prognostic marker studies are published each year. This review provides an overview of current biomarkers in ovarian cancer and their potential utility in clinical practice as prognostic biomarkers (i.e., those that can be used to estimate the chances of disease recurrence) and predictive markers (ie, those associated with response or lack of response to a particular therapy). A summary of these biomarkers is provided in Table 1.

Table 1.

Prognostic Biomarkers of Epithelial Ovarian Cancer

Biomarkers Clinical Utility
Classic plasma/serum markers
CA 125 Postoperative levels of CA 125 >35 U/mL (no residual) or >65 U/mL (residual) are independent prognostic factors for survival14
HE4 High serum level predicts unfavorable prognosis, HE4 combined with CA(125) assay can improve diagnosis272
A change in HE4 level of ≥25% is considered significant8 (an increase of this magnitude suggests recurrence or disease progression; a decrease suggests therapeutic response)
Mesothelin High mesothelin expression is associated with chemoresistance and with shorter disease-free survival and worse overall survival.273
M-CSF Markedly elevated levels of M-CSF1 in serum and ascites are associated with a poor prognosis274
Osteopontin (OPN) Osteopontin level in metastatic lesion may be a prognostic indicator in ovarian cancers275
Expression of kallikrein (KLK)-related peptidases Several kallikrein-related peptidases (such as KLK6, KLK13), in addition to CA125, can provide a significant advantage to detect early-stage ovarian cancer276
Preoperative high plasma bikunin level There is a 2.2 fold increased risk of death for patients with lower plasma bikunin levels277
Novel plasma/serum markers
Plasma cell-free DNA Tumor-specific CFDNA levels correlate with increasing tumor burden and decline following therapy151
Angiogenic markers
VEGFs Heterozygous genotype in VEGF +405G/C or VEGF -460C/T; higher serum expression of VEGF is associated with a shorter overall survival; elevated VEGF ascites levels negatively correlate with patient survival278, 279
VEGF +405G/C, VEGF -460C/T, and VEGF +936C/T do not correlate with any of the investigated clinicopathological parameters280
EphA2 expression Overexpression is associated with poor prognosis46
FGF-1 Overexpression of FGF-1 is significantly associated with overall survival281
EZH2 Overexpression of EZH2 promotes the proliferation and invasion of human EOC cells282
Claudin family members Claudin-3 and claudin-7 expression in effusions independently predicts poor survival283
Apoptotic /signaling pathway and a genetic related markers
EGFR and HER2 Tumors with increased EGFR protein tend to grow more aggressively, are more likely to metastasize, and are more resistant to chemotherapy284; HER2 overexpression is associated with poor overall survival285; HER-2/neu expression does not appear to be an important prognostic factor in patients with advanced epithelial ovarian cancer286
p53 mutation p53 antibodies have shown mixed results with regard to prognostic value287,288
Cyclin D1, cyclin E Overexpression of cyclin D1 is significantly related to a more aggressive tumor phenotype and poor prognosis in ovarian carcinoma289
p16-cyclin D1/CDK4-pRb pathway is a prognostic factor for patients with abnormal G1 pathway.310
Overexpression of cyclin E is associated with poor outcome 187
Serum sFas levels Survival rates decrease as serum sFas levels increase311; Serum sFas level is also a useful biomarker for predicting response to platinum-based chemotherapy312
ERCC1 ERCC1 protein overexpression may act as a prognostic marker for poor survival of high-grade OC patients and as indicator for debulking outcome in advanced EOC290292
Low ERCC1 expression correlates with improved survival in advanced OC212
BRCA1/2 carriers The BRCAness profile correlates with responsiveness to platinum and PARP inhibitors293
Patients with BRCA1-2 mutations represent only a subgroup within the group of patients with double primary breast and ovarian cancer294
Low BRCA1 expression correlates with improved survival in advanced OC212
Immune-related markers
IL-6, IL-7, IL – 8 Elevated IL-6 serum levels correlate with poor prognosis55
Increased IL-6 and IL-8 expression correlates with poor initial response to paclitaxel chemotherapy; elevated expression of IL-6 correlates with poor final outcome295
IL-7 levels are strongly associated with ovarian cancer and might be used in combination with CA-125 to distinguish between malignant and benign ovarian tumors296
Ascitic-fluid IL-12 levels High ascitic-fluid IL-12 levels are associated with disease progression297
APM (antigen processing machinery) component APM (antigen processing machinery) component (TAP1, TAP2, HLA-HC, and beta 2 microglobulin) downregulation is significantly associated with improved survival220
B7-H3 B7-H3 expression in tumor vasculature may reflect tumor aggressiveness298
B7-H4 High B7-H4 levels correlate with poor prognosis; however, the effect is not significant when outcome adjusted for other clinicopathological variables299
Intratumoral CD3+, CD8+ T cells infiltration Intratumoral infiltration of CD3+ or CD8+ T-cells is significantly associated with improved survival220
Gamma - interferon expression Elevated interferon-gamma expression correlates with improved clinical outcome in patients with ovarian cancer300
Loss of IFN gamma receptor independently predicts poor prognosis301
Tissue-based markers
Claudin-3 High claudin-3 expression is associated with shorter survival
Claudin-4 Claudin-4 overexpression in epithelial OC does not correlate with survival or other clinical endpoints
MMP-2, MMP-9, and MT1-MMP High epithelial and stromal expression of MMP-2, MMP-9, and MT1-MMP are each significantly associated with shorter disease-specific survival and are poor independent predictors of survival
FAK Overexpression is associated with significantly worse survival
Levels of miR-200, miR-141,miR-18a, miR-93, miR-429, let-7b, miR-199a Low-level expression of miR-200 miRNA predicts poor survival.302
Higher expression of miR-200, miR-141, miR-18a, miR-93, and miR-429, and lower expression of let-7b, and miR-199a correlates significantly with poor prognosis232
Dicer, Drosha expression Dicer and Drosha mRNA levels in ovarian-cancers is associated with patient survival238
Decreased Dicer expression correlates significantly with reduced patient survival in serous cancers and advanced disease240

2. Classic Biomarkers of Epithelial Ovarian Cancer

2.1. Clinical utility of CA125

CA125, the first ovarian cancer biomarker ever identified, is a high-molecular-weight glycoprotein recognized by the monoclonal antibody OC125. CA125 is expressed by epithelial ovarian tumors and other tissues of Mullerian origin. Serum CA125 levels are initially elevated in >80% of ovarian cancer patients and may be used to reflect the clinical course of disease.4 Since Bast and colleagues first produced the OC125 monoclonal antibody in 1981,5 serum CA125 measurement has been used extensively in clinical practice to screen women at high risk for ovarian cancer, to plan treatment, and to predict clinical course and response to chemotherapy. Despite having limited sensitivity and specificity, serum CA125 estimation is generally agreed to have clinical value during preoperative work-up and during post-treatment monitoring.4, 6, 7 Serum CA125 is also of prognostic significance in predicting patient survival and response to chemotherapy.4, 810 Therefore, CA125 remains one of the most important prognostic biomarkers for EOC patients in the clinical setting.911

It is commonly agreed that patients with early-stage EOC are less likely to have an elevated serum CA125 level preoperatively and more likely to have a better prognosis.4, 6, 7, 12 One previous study showed a significantly higher survival rate for patients with preoperative serum CA125 <65 U/ml than for patients with preoperative serum levels >65 U/mL.13 Postoperative serum CA125 >65 U/mL was also associated with a significantly worse 5 year survival than was postoperative serum CA125 < 65 U/mL (42% versus 5%). Makar and colleagues found that elevated serum CA125 was an independent prognostic factor for survival, regardless of residual disease status after primary surgery.14

2.1.1. CA125 as prognostic factor

In patients without residual disease after primary surgery, significant prognostic factors for survival include histology type, postoperative CA125 level with a cutoff value 35 U/mL, and tumor grade ; for those with residual disease, they are histology type, postoperative CA125 level with cutoff value 65 U/mL, postoperative treatment, and size of residual disease. Dynamic change in serum CA125 level before and after treatment is also an independent prognostic factor for survival.14, 15 In a 10–year European study, Han and collaborators recruited 296 patients who met the criteria of CA125 progression and doubling during surveillance after first-line therapy. During surveillance, the median CA125 doubling time was 40 d; the median survival was shorter for patients whose CA125 doubling time was ≤ 40 d than for those whose doubling time was > 40 d (10.6 months versus 22.1 months). On multivariate analysis, short CA125 doubling time (≤ 40 d) and short time to disease progression (6180 d) remained the only 2 independent adverse prognostic factors. Han et al also confirmed these findings in a set of 28 new patients. Together, their findings suggested that the rate of CA125 increase during surveillance has independent prognostic significance.13 Therefore, serum CA125 level should be considered when making therapeutic decisions for EOC patients.

2.1.2. CA125 as chemoresponsiveness predictor

Dynamic changes in serum CA125 levels can also be used to predict response to first-line and second-line chemotherapy.9, 1621 This enables clinicians to plan primary treatment and to make the most appropriate second-line treatment decisions for patients with EOC. Some investigators have proposed using serum CA125 as a marker in clinical trials of new therapeutic drugs for ovarian cancer,22 which is now the case in new EOC drug development.23

Karam et al proposed that withholding treatment in the event of isolated rising CA125 levels would not negatively affect these patients’ overall survival,24 thereby highlighting the need for more efficient clinical use of serum CA125 testing and for new EOC biomarkers. Clinical and basic research aimed at overcoming the drawbacks of CA125 by means of (1) dynamic CA125 monitoring, (2) combination with ultrasonography and other clinical factors such as TNM and stage, and (3) combination with other biomarkers has produced encouraging results.11, 15, 2426

3. Novel biomarker development in EOC

Thanks to the burgeoning development of genomics, proteomics, metabolomics, and other “-omics”’ and an improved understanding of cancer biology over the past 3 decades, hundreds of prognostically valuable EOC biomarkers have been discovered. Some of the most promising of these biomarkers are VEGF, HE4, mesothelin, M-CSF, osteopontin, kallikrein(s), bikunin, EphA2, and soluble EGF receptor, although none surpass CA125.11, 25, 2735 Those most relevant to the recent clinical focus on targeted and personalized therapy are biomarkers of angiogenesis and related signaling pathways.

3.1. EOC biomarkers with angiogenic features

It is well established that angiogenesis (ie, the formation of new blood vessels) is necessary for the growth and metastasis of solid tumors.36 Tumor angiogenesis is regulated by a complex, and still not fully understood, interplay between endogenous angiogenic stimulators (such as VEGF and FGF family members) and inhibitors (such as endostatin and thrombospondin-1).37 Angiogenic biomarkers have been the subject of recent comprehensive reviews. Alterations in the expression of pro- and anti-angiogenic factors serve not only as markers of solid tumor progression but also as prognostic indicators for EOC.2833, 35, 3840

VEGF was the first, and is now the most extensively studied, family of angiogenic factors. VEGF, the most prominent of the angiogenic stimulators, exists in four main forms of 121, 165, 189 and 206 amino acids in length. Each has a variety of functions, including recruitment and mitogenic stimulation of endothelial cells.41 In a study of VEGF expression in tumor specimens from 68 FIGO stage I and II EOC patients, median disease free survival was better in VEGF negative than VEGF positive patients (18 months vs. 120 months), and elevated VEGF expression was associated with poorer survival.29 Serum VEGF elevation pretreatment, thought by some to be a good predictor of tumor angiogenesis, correlates significantly with poorer disease-free survival and overall survival.32 VEGF-A and other VEGF family members such as VEGF-D are independent prognostic factors in EOC.38 Other researchers have reported similar results.28, 30, 31, 33, 35, 39, 42 Because VEGF is one of the more prominent angiogenic mediators of new tumor vessel growth and also an important endothelial cell survival factor,43 it is both a potential biomarker and a potential therapeutic target. Indeed, VEGF targeted therapy has shown clinical promise,44, 45 but its therapeutic effect is largely limited to newly forming tumor vessels and is dependent on complicated interactions with other factors such as EGF, PDGF, IL-8, EphA2, and the matrix metalloproteinases. Meanwhile, there is a lack of effective biomarkers for monitoring response to new antivascular agents.

EphA2, a transmembrane receptor tyrosine kinase overexpressed by many human cancers, is another promising angiogenesis-related prognostic biomarker.28, 46, 47 It is often associated with poor prognostic features and tumor angiogenesis.46, 47 EphA2 is a member of the largest family of tyrosine kinase receptors The EphA2 family consists of 16 known receptors and 9 known ligands that are categorized into subtypes A and B by the ephrins with which they interact. EphA2 is involved in many processes crucial to malignant progression, such as migration, invasion, metastasis, proliferation, survival, and angiogenesis. It is overexpressed in cancers of the ovary, breast, prostate, lung, colon, esophagus, renal cell, cervix, and melanoma. In many of these malignancies, EphA2 overexpression is associated with poor survival, advanced stage, or increased metastatic potential. In an immunohistochemical study of 94 tissue specimens (EOC and control) by our group, EphA2 overexpression was significantly associated not only with higher tumor grade and advanced stage of disease, but also with significantly shorter survival (median, 3.1 years vs. 12 years for patients with low or absent EphA2 tumor expression), even after multivariate analysis in a Cox proportional hazards model.28 Another study in endothelial and ovarian cancer cells revealed a strong association between EphA2 overexpression and the expression of critical tumor angiogenic and invasive factors.47

Given the positive association of EphA2 with aggressive clinical and pathological features in human cancers and the cancer-promoting features of EphA2 overexpression,28 we have examined the therapeutic potential of downregulating EphA2 expression in preclinical models. We found that silencing EphA2 alone could reduce tumor growth by up to 60% and, in combination with antibody/paclitaxel therapy, by up to 95%.46, 4850

Our recent study of the enhancer of Zeste homolog 2 (EZH2) showed the functional and biological importance of EZH2 in angiogenesis and tumor growth and the potential value of increased EZH2 expression in either tumor cells or tumor vasculature as a predictor of poor clinical outcome. This suggests the therapeutic potential of targeting EZH2.51 Many other angiogenic factors have potential prognostic value in EOC, yet, their prognostic significance has to be confirmed in further studies. These factors include FGF family members, interleukins (especially IL- 6 and IL-8), cell-cell and cell-ECM (extracellular matrix) factors, including several claudin family members (claudin-3, claudin-4, and claudin-7) and matrix metalloproteinases (MMPs) (eg, MMP2 and MMP9), focal adhesion kinase (FAK), and tissue factor (TF), and their alterations in tissue, serum, and other biological fluids including cyst fluid and ascites. 5153

Given the role of cytokines in the biology of ovarian cancer cells, the current interest in their diagnostic, monitory, and prognostic uses is not surprising. Interleukins are potent endothelial-cell chemo-attractants. Current investigations have focused on IL-6 as a prognostic factor. High IL-6 levels have been associated with unfavorable clinical outcome,5456 though not always consistently.57 IL-6 level reflects a large tumor burden, and macrophage activation followed by subsequent release and accumulation of IL-6 in the serum and ascites and the expression of factors that might affect tumor burden and macrophage activation are not easy to control. In one study, IL-6 was no better than traditional markers as a disease biomarker when used alone, but was considered useful when used as part of a panel of cytokines.58 Although IL-6-mediated tumor growth and angiogenesis has been demonstrated in ovarian cancer models,59 the benefit of IL-6 as a marker of ovarian tumor angiogenesis remains to be determined.52

Interleukin-8, one of the most widely studied cytokines in ovarian cancer, is a potent proangiogenic factor overexpressed in most human cancers, including ovarian carcinoma.6063 Because IL-8 helps mediate tumor angiogenesis,63 quantifying circulating IL-8 levels may be a useful biomarker of response. High IL-8 expression has been associated with advanced tumor stage, high tumor grade, and worse survival (median survival for patients with high vs. low IL-8 expression: 1.62 vs. 3.79 years). Increased IL-8 expression has also been associated with poor clinical outcome in human ovarian carcinoma and IL-8 gene silencing with decreased tumor growth via anti-angiogenic mechanisms.64

Conversely, others have reported that IL-8 levels increase immediately after the initiation of chemotherapy in ovarian cancer patients, especially in those with residual disease.65 This reflects not only the secretion of IL-8 by multiple sources including monocytes, neutrophils, and mesothelial cells, but also the need for further preclinical investigations of IL-8 as a tumor biomarker. The claudins are integral components of tight junctions (ie, apical cell-cell adhesions that regulate epithelial paracellular permeability and are critical for epithelial cell polarity). In tumor cells, the loss of cell-cell adhesion is central to cellular transformation and metastatic potential.66, 67 Certain members of the claudin gene family including claudin-3, -4, and 7 are abnormally regulated in several human cancers including ovarian cancer.68, 69 Claudin-3 is overexpressed in 90% of ovarian tumors, including all 4 major subtypes (serous, mucinous, clear cell, and endometrioid),70 and is also associated with shorter survival.71 Claudin-3 expression also promotes migration, invasion, and survival of ovarian cancer cells.68, 72 Conversely, silencing the claudin-3 gene suppresses ovarian tumor growth and metastasis in mouse tumor models.70 Claudin-4 overexpression in EOC apparently does not correlate with survival or other clinical endpoints.69

Tissue MMPs are a large family of zinc- and calcium-dependent proteolytic enzymes that can degrade most components of the extracellular matrix and mediate tumor invasion and metastasis.73 The MMPs associated with ovarian carcinomas include MMP-2, MMP-9, and membrane type-1 (MT1)-MMP according to majority of authors.7478 MMP-2, MMP-7, MMP-8, MMP-9, and MT1-MMP have been detected in 54%, 81%, 95%, 97% and 100% of EOCs, respectively.78, 79 Torng et al found stromal MMP-2 expression to be a poor predictor of disease specific survival for patients with endometrioid cancer, but not for those with serous histology.80 Davidson et al found MMP-9 mRNA expression in tumor cells to be an independent poor prognostic predictor of survival in patients with EOC.81 Others have reported that patients with weak tumor expression of stromal MMP-9 had significantly longer survival than did those with moderate or intense expression.82 In a study of 90 EOC tissue specimens, Kamet et al reported that high epithelial and stromal expression of MMP-2, MMP-9, and MT1-MMP were each significantly associated with shorter disease-specific survival on univariate analysis, and that high stromal expression of MMP-9 and MT1-MMP and strong epithelial expression of MT1-MMP were poor independent predictors on multivariate analysis.78 In contrast, Westerlund et al reported that, stromal MMP-2 expression correlated with earlier ovarian carcinoma and better prognosis.83

Cell migration is an important component of the metastatic process and requires repeated adhesion to and detachment from the extracellular matrix microenvironment. These events are mediated, in large part, by integrins. FAK, an integral component of the integrin-signaling pathway, is a critical mediator of signaling events between cells and their extracellular matrix, thereby facilitating invasion and migration.84 FAK is overexpressed in metastatic human colorectal, breast, thyroid, and prostate cancer cells and ovarian cancer.8589 Its overexpression predicts poor clinical outcome by virtue of its significant association with worse survival (median, 7.6 years versus 2.98 years).90 In a series of in vitro and in vivo experiments in which ovarian cancer cells were transfected with the dominant-negative construct FRNK, FAK inhibitor-TAE226, or FAK-siRNA-DOPC separately, the inhibition or silencing of FAK and its phosphorylation significantly reduced tumor burden in multiple tumor models and decreased invasion, migration, and cell spread.40, 90, 91 Further studies are warranted and necessary to translate these experimental findings into clinical practice. Other angiogenic markers, such as tumor cell–lined vasculature, microvessel density (MVD), and stromal compartment, are potentially prognostic and therefore potentially therapeutic in EOC.31, 42, 47, 92, 93 However, their utility after anti-angiogenic therapy remains to be determined. Consequently, we consider them to be microscopic level markers and outside the scope of this review.

Modern gene array-based profiling approaches have been used to develop EOC angiogenic biomarkers.94, 95 Using immunohistochemistry-guided laser-capture microdissection and genome-wide transcriptional profiling techniques, Buckanovich et al identified and validated 12 novel ovarian tumor vascular markers (TVMs) and found that overexpression of any one of three ovarian TVMs by vascular cells was associated with decreased disease-free interval. Using a similar approach, Mendiola et al tested 61 formalin-fixed, paraffin-embedded samples from late stage I EOC patients and generated a 34-gene profile that had independent prognostic value on multivariate analysis. This gene profiling approach will be discussed in more detail later in this review. 95

Despite the promise of anti-angiogenic therapy for EOC, its potential clinical use remains controversial.27, 9698 Several of the most promising anti-angiogenic drugs, which target VEGF and VEGF related genes (eg, bevacizumab, sorafenib, sunitinib, and temsirolimus), are already approved by the FDA for use against solid tumors, such as advanced RCC and metastatic colorectal cancer, but their use in EOC remains the focus of preclinical and clinical trials.96 Nevertheless, the promising results achieved with angiogenesis-related factors over the last few decades may soon lead to the targeted application of such factors in EOC and their use as biomarkers of clinical course and responsiveness.

3.2. Signaling pathway, apoptotic pathway, and genetic/ epigenetic related markers

Successful biomarker development depends on approaches that originate from the discovery phase and culminate in the clinical validation of an appropriately targeted biomarker. The most frequently reported molecular pathway biomarkers in ovarian cancer are EGF receptor (HER) family members, apoptotic pathway surrogates (such as p53 and cell cycle- related kinases) and DNA repair proteins (such as BRCA1/2 and ERCC1).

3.2.1. EGF receptor (HER) family

The human EGF receptor (HER) family mediates crucial cellular processes, including growth, proliferation, and survival. This receptor family consists of four transmembrane receptors: HER1 (EGFR), HER2, HER3, and HER4. Each receptor has an extracellular binding domain, a transmembrane domain, and an intracellular domain.99, 100 This receptor is noted for the interdependence and functional complementarity of its members and their tendency to heterodimerize with each other (eg, HER2/HER3, HER2/HER4), traits that have significant implications for the development of targeted therapies.101, 102 EGFR (HER) – 1,2 and 4 proteins possess tyrosine kinase activities, whereas EGFR(HER)–3 doesn’t, but EGFR(HER3)-3 can dimerize with other EGFR(HER) family members and lead to activation of the EGFR(HER) pathways.103

EGFR is widely expressed in a variety of human tumors including ovarian, head and neck, breast, and non-small-cell lung (NSCLC) cancers.104 Estimates ranging from 60% to 98% of all epithelial ovarian cancers express high levels of the EGFR (HER), and extensive studies also revealed that EGFR (HER) had significant implication in tumor growth and progression.103, 105108 Two separate groups have shown an inverse correlation between EGFR and survival in ovarian cancer.109, 110 Others have delineated the key role of EGFR in downstream signaling pathways such as the PI3K (phosphatidylinositol-3 kinase)/Akt and ERK (external signal-regulated kinase) pathways.111 Still others have revealed a positive correlation between nuclear EGFR and cyclin D1 and K1–67.112

The role of HER2 has been studied in several tumor types, including breast, head and neck, prostate, and ovarian cancer.113 In breast cancer, HER2 overexpression varies widely (from 1.8% to 76%).114 In a randomized trial, Slamon found HER2 expression in breast and ovarian carcinoma to be indicative of aggressive tumor growth and poor prognosis.115 In a Dutch study of 208 patients, mean survival was shorter in patients with HER2-positive tumors than in those with HER-2 negative tumors (17 months vs. 26 months).116 Similar findings were made by Tanner et al in a study of 96 women with stage III ovarian cancer (12 months vs 25 months)117 and by Felip et al (62 weeks vs. 123 weeks).118 Other studies have shown HER2 expression to be an independent prognostic indicator of survival.119, 120

In a study of patients with HER2-negative ovarian tumors, Gorden et al found that HER2 was activated in 45% of the tumors and that HER2 amplification was not required for any clinical activity associated with pertuzumab.121 Others have shown that HER2 may activate key downstream signaling pathways (eg, PI3K—Akt and RAS-Raf-MEK) by dimerizing with other ligand-activated HER receptors, particularly HER3.122124 Indeed, HER2 preferentially binds with HER3, and the resulting HER2/HER3 heterodimer has a strong mitogenic signaling effect.125 In a study of 116 consecutive patients with primary EOC, HER3 levels were associated with patient survival on both univariate and multivariate analysis.126 Moreover, as shown by Kaplan-Meier analysis, patients with low HER3 protein levels (n = 54) had a significantly longer median survival time than did patients with high HER3 protein levels (n = 62) (3.31 years versus 1.80 years). HER3, lacking of tyrosine kinase activity itself, however, after dimerization with other members of the EGFR family, several signal transduction cascades can be activated, including PI3-K (phosphoinosite 3’-kinase)/Akt and extracellular signal-regulated kinase (ERK1/2). In this study, they reported that HER3 overexpression was observed in 53.4% of patients.126

HER2 and HER3, by themselves, are functionally incomplete receptors and therefore particularly dependent on each other. HER2 has an extracellular domain but no apparent ligand-binding activity, while HER3 has a non-functional kinase domain and no catalytic kinase activity. In contrast, the HER2/HER3 heterodimer is a highly functional signaling unit and is in fact the most active signaling dimer in the HER family.127 HER4 mediates antiproliferative effects. However, its overactivity does not appear to play a major role in tumorigenesis.128 Moreover, there exists the debate on the prognostic value of EGFR (HER) in EOC. Graeff and colleagues performed a meta-analysis on total 15 EGFR studies, 20 HER-2 /neu studies and 62 p53 studies; they concluded that although p53, EGFR and HER-2/neu status modestly influences survival, these markers are, by themselves, unlikely to be useful as prognostic markers in clinical practice. Thus, further validation based on studies with well-defined, prospective clinical trials and more complete reporting of results are needed.129

Biologically targeted antibodies and tyrosine kinase inhibitors directed toward EGFR/ErbB1/HER1 (eg, cetuximab, erlotinib and gefitinib) and ErbB2/HER2 (eg, trastuzumab), and more recently toward ErbB3/HER3 and ErbB4/HER4, are being investigated as therapies for patients with EGFR/ERBB/HER proto-oncogene-driven malignancies.130 Ongoing trials of these agents alone or in combination with traditional chemotherapeutic agents are underway.131 Trastuzumab, a monoclonal antibody against HER2, is the standard of care for patients with HER2-positive breast cancer in the metastatic and adjuvant settings and is associated with improved disease-free and overall survival. However, in a phase II trial of single-agent trastuzumab in patients with persistent or recurrent epithelial ovarian or primary peritoneal cancer, no relationship was found between HER2 expression and clinical response, progression-free survival, or overall survival.132 Interestingly, the combination of cytotoxic chemotherapy and EGFR/HER inhibitors seems to provide better clinical response and therefore warrants further evaluation. Inhibition of HER2/HER3 dimerization is a particularly attractive approach to anticancer therapy.124, 133

3.2.2. Apoptotic pathway biomarkers

3.2.2.a. p53

The p53 suppressor gene has come to the forefront of cancer research because it is commonly mutated in human cancer. Thus, detection of p53 abnormalities may have diagnostic, prognostic, and therapeutic implications. p53 is mutated in all major cancers,134 and represents the most frequent mutation in ovarian cancer.135 p53 is one of the most important mediators of the intrinsic apoptotic pathway. Upon activation, the p53 protein acts as a tumor suppressor whose genetic effects lead to cell-cycle arrest, apoptosis, senescence, or differentiation.136138 p53 is mutated in 60–80% of all sporadic EOCs and correlates highly with high-grade tumors.139143 Subtype-specific mutations in p53 have been found in almost 60% of serous tumors but only 16% of mucinous tumors,144 which suggests a prominent role for p53 mutation in carcinogenesis of serous but not mucinous ovarian tumors. p53 mutation is a late event in ovarian carcinogenesis, and the loss of p53 apparently confers a more aggressive, rapidly growing phenotype. Several studies have examined the prognostic value of p53 antibodies in ovarian cancer.145 One study on total 126 patients reported that the plasma p53-Ab positivity was correlated with the highest risk of cancer progression in serous EOC.146 In another study of presurgical serum from 104 women with ovarian cancer, p53 antibody was associated with better overall survival. In a meta-analysis, patients with aberrant p53, EGFR, or HER2 tumor status had significantly lower odds of surviving 5 years. Moreover, Goodell and colleagues evaluated the relationship between antibody immunity to the p53 oncogenic protein with EOC outcome, and they demonstrated the presence of p53-Ab was an independent prognostic indicator of overall survival in advanced-stage patients.147

Other studies have associated overexpression of mutant p53 with worse survival, FIGO stages,148151 poorer histological grade of differentiation, and high proliferative fraction. Shahin et al observed an association between worse prognosis and p53 mutation status in patients with EOC. Overall, 48.5% and 57.3% of tumor samples from those patients showed p53 overexpression and p53 mutation, respectively; and even though neither p53 overexpression nor p53 mutation affected overall survival, the combination did predict worse survival in both univariate and multivariate models. When p53-nonresponsive and p53-null tumors were grouped together as functionally null, the worst prognosis was associated with missense mutations on univariate analysis. Functionally null p53, stage, and optimal cytoreduction were independent prognostic factors on multivariate analysis. More detailed characterization of p53’s mutational spectrum and classification of mutations according to their effect on p53 functionality have led some groups to hypothesize that distinct types of mutations can shorten survival.152, 153 However, there is no consensus to date, and regardless of tumor stage, the effect of p53 mutation status on overall survival remains questionable. Some studies have found no association between any p53 alteration and prognosis.154156

3.2.2.b. Cell cycle-related kinases

Cell cycle deregulation is a hallmark of carcinoma.37 CCRK (cycle-related kinase) is frequently overexpressed in brain, renal cell, and ovarian cancers and is expressed either weakly or not at all in normal ovarian epithelial tissue.157, 158 In a study by Wu et al, overexpression of CCRK in ovarian carcinomas correlated strongly with ascending histopathological grade, poor differentiation, and/or advanced clinical stage and thus served as an independent predictor of shorter overall survival. Upregulated expression of CCRK in ovarian carcinomas might involve the cyclin D1-associated pathway. Cyclin D1 is essential for G1 phase progression and is a candidate proto-oncogene.159 Cyclin D1 overexpression is associated with tumor characteristics and clinical outcome in many cancers including ovarian cancer.160166 In a study of 134 patients with serous EOC,167 cyclin D1 overexpression (>10% versus ≤10%) was an independent predictor of poor survival. Such findings, however, are not fully consistent with those by other groups.168, 169 In a Cox regression analysis, Barbieri et al found that women with cyclin D1-overexpressing ovarian adenocarcinomas and residual disease > 2 cm after surgery had significantly shorter survival than those who did not (relative risk(RR) of death: 2.48 and 3.7, respectively). These investigators did not, however, note any significant difference in cyclin D1 overexpression between histologic subtypes.168 On the other hand, Masciullo et al165 found that overexpression of cyclin D1 mRNA in EOC was significantly associated with well-differentiated and moderately well-differentiated tumor grade (grade 1–2) but not with clinical outcome and other parameters. Moreover, no association has been found between the presence of cyclin D1 expression and clinical outcome.161, 164, 168 These contrasting results are likely due to differences in analytical techniques and scoring systems used in those studies.170

The role of cyclin E in oncogenesis and its function as a clinical prognostic indicator in cancer patients have been the focus of many reports in recent years. Cyclin E has been consistently associated with disease progression in various malignancies and with poor prognosis in patients with breast, bladder, colorectal, and ovarian carcinoma.171178 Cyclin E gene amplification has been detected in 12% to 21% of ovarian tumors,177179 RNA overexpression in up to 30% cases,177179 and protein overexpression in up to 70%.180182 Together, these findings suggest that cyclin E is an important mediator of survival and its expression an independent predictor of poor prognosis in patients with ovarian carcinoma. Overexpression of the cyclin E protein has been linked to shortening of the G1 phase of the cell cycle, decreased requirement for growth factors, enhanced cell proliferation, induction of chromosomal instability, and polyploidy.172, 183186 These processes contribute to the oncogenic potential of cyclin E. Cyclin E’s prognostic value is debatable. Rosen et al demonstrated that a high level of cyclin E is an independent predictor of poor prognosis.187 Sui et al correlated increased cyclin E expression with tumor progression and poor prognosis in EOC patients.180 In contrast, others have found no correlation between cyclin E expression and survival in patients with ovarian carcinoma 167, 188 or only a trend toward an association with a poor prognosis.189

3.2.3. BRCA1 and BRCA2

The BRCA1 and BRCA2 tumor suppressor genes help regulate cellular proliferation, chromosomal stability, and DNA repair via homologous recombination. They help maintain genomic stability by recognizing and mediating repair of DNA damage, regulating transcription, and controlling cell cycle checkpoints through p53-dependent and independent mechanisms.190192 Approximately 10% of breast and ovarian carcinomas are due to a genetic predisposition (mainly germline mutations in the BRCA1 and BRCA2 genes).190 Females carrying BRCA1 or BRCA2 mutations have a 16%–60% and a 16%-27% lifetime risk, respectively, of developing EOC.190, 193 Studies in high-risk families indicate that mutations in BRCA1 or BRCA2 genes confer as much as an 87% risk of breast cancer and 44% risk of ovarian cancer by the age of 70 (with mean age of onset 5–10 years earlier than in non-carriers) and that the vast majority of these patients will have serous adenocarcinomas.194196 Interestingly, however, most studies indicate that women with a BRCA1 or 2-related ovarian cancer have better survival than non-carriers, 197204 particularly if they receive platinum-based therapy. 201204 In vitro studies have shown that some BRCA1 mutated ovarian cell lines have increased sensitivity to various chemotherapeutic agents.171 These findings are supported by an unbiased historical cohort study of Ashkenazi Jews in which ovarian cancer outcomes were better for BRCA1 or 2 mutation carriers than for non-carriers199 and by studies showing a more favorable response to chemotherapy among patients with hereditary ovarian cancer.199, 200, 205, 206

3.2.4. ERCC

ERCC1 (excision repair cross-complementation group 1) is a major mediator of nucleotide excision repair (NER), a process that can correct most bulky lesions in DNA.207 ERCC1 overexpression is a potential marker of prognosis and of tumor resistance to platinum-based chemotherapy regimens in many cancers, including EOC.207214 In patients with advanced EOC, higher levels of serum ERCC1 protein are often associated with widespread metastasis, enhanced DNA damage repair, and tumor aggressiveness. On the contrary, lower levels of ERCC1 (and resulting lower rates of DNA repair and increased early senescence and apoptosis of tumor cells) are often associated with slower metastasis and fewer lesions, which might render such tumors more amenable to optimal debulking surgery.211 Studies in EOC patients support this view. ERCC1 was shown to be an independent prognostic marker of debulking outcome, and ERCC protein expression was associated with survival or response to platinum compounds (but not always with debulking outcome).215, 216

3.3. Epithelial ovarian cancer biomarkers with immune signatures

Given the well-established importance of the immune system in tumor development,217, 218 researchers are striving to find novel immune-related prognostic biomarkers. Zhang and colleagues reported that the presence of intratumoral T cells correlated with improved clinical outcome in advanced ovarian carcinoma from a study on 186 ovarian cancer specimens. They found that the five-year overall survival rate was 38% in patients with positive intratumoral T cells, compared with 4.5% in patients without T cells in tumor islets. They also found significant differences in progression-free survival and overall survival according to the presence or absence of intratumoral T cells in 74 patients with a complete clinical response after debulking and platinum-based chemotherapy (the five-year overall survival rate: 73.9% vs. 11.9%). The presence of intratumoral T cells independently correlated with delayed recurrence or delayed death on multivariate analysis and was associated with increased intratumoral expression of interferon, interleukin-2, and lymphocyte-attracting chemokines. The absence of intratumoral T cells was associated with increased levels of vascular endothelial growth factor.219

Several novel immune-related biomarkers of EOC have independent prognostic value. One of these is the antigen processing machinery (APM) components. Han and colleagues were the first to report that APM downregulation and lack of intratumoral T-cell infiltrates are independent prognostic markers for death from ovarian carcinoma. In their study of 150 tumor samples from patients with invasive EOC, they found that most tumors expressed TAP1, TAP2, HLA-HC, and β2 microglobulin either heterogeneously or positively; 67% possessed intratumoral CD3+ or CD8+ cells; and 88% possessed peritumoral CD3+ or CD8+ cells.220 Another prognostically valuable immune-related biomarker is B7-H3. Recently, Zang et al reported that B7-H3 (one of the B7 family members of immunoregulatory ligands), positive tumor vasculature was associated with significantly shorter survival time and a higher incidence of recurrence.221 Together, these findings imply the critical role of immune surveillance and immune escape in the clinical course of EOC.

3.4. MicroRNA, Dicer, and Drosha --- EOC biomarkers on the frontier of discovery

Recently, a new epigenetic regulatory program was identified with the discovery of small non-coding microRNAs (miRNAs) in Caenorhabditis elegans.222 MiRNAs are endogenous, small non-coding RNAs (approximately 22 nucleotides) that negatively regulate gene expression at the posttranscriptional level in a sequence-specific manner.223226 miRNAs may play a role in cancer development and progression. For instance, loss of miRNA-mediated gene regulation may be a tumorigenic mechanism,223 impaired miRNA processing may enhance cellular transformation and tumourigenesis,227 and miRNAs may act as oncogenes or tumor suppressor genes.228, 229 These insights have stimulated investigations into miRNA’s role in ovarian cancer. An early study showed that approximately 40% of miRNA genes exhibit altered DNA copy numbers.230 More recent studies have shown that deregulation of miRNAs affects survival and drug resistance.226, 231, 232 In a study by Nam et al, higher expression of miR-200, miR-141, miR-18a, miR-93, and miR-429, and lower expression of let-7b, and miR-199a correlated significantly with poor prognosis.246 In a study by Yang et al, miR-214, miR-199*, and miR-200a were associated with high-grade and late-stage ovarian tumors.63 Other studies have shown that miRNAs may be useful in predicting ovarian carcinoma outcome.233 For example, many members of the let-7 protein family act as tumor suppressors, and patients with low let-7a-3 methylation levels have worse overall survival than those with high methylation levels.233 In addition, even though most miRNAs are downregulated in cancer tissues, some are upregulated in recurrent as opposed to primary tumors.

Increased knowledge of the RNA-interference machinery and its 2 main components—the miRNAs Dicer and Drosha—has led to investigations of their use as diagnostic, prognostic, and therapeutic biomarkers in cancer.234238 In mammalian miRNA biogenesis, the primary transcripts of miRNA genes (pri-miRNAs) are cleaved into hairpin intermediates (pre-miRNAs) by Drosha (nuclear RNase III) and further processed into mature miRNAs by cytosolic Dicer (another RNase III-related enzyme).233 Dysregulation of miRNA secondary to the defects in RNA silencing machinery has been observed in several types of tumors including ovarian cancers.238, 239 We were the first to report that low Dicer expression independently predicted poor outcomes in ovarian cancer patients.238 In line with our results, Faggad et al provided evidence that the decreased expression of Dicer has functional consequences for miRNA expression and leads to a global downregulation of miRNA expression and significant changes in gene expression.240 When we extended our analysis to other tumor types using mRNA expression profiling data, we confirmed a similar relationship between Dicer expression and outcomes in lung, breast, and other cancers 239, 241 Dicer expression appears to be upregulated in non-invasive precursors of invasive lung adenocarcinoma. 239 Contrary findings in other tumor types have been reported (ie, correlation of high Dicer and Drosha expression with poor prognosis in prostate cancer239 and no correlation of Dicer expression with outcome in acute myeloid leukemia),242 although these discrepancies might be explained by tissue specificity.

Suzuki et al have forged a link between the tumor suppressor p53 and posttranscriptional maturation of miRNAs by demonstrating that p53 interacts with the Drosha microprocessor complex through DEAD-box RNA helicase p68 (DDX5) to facilitate the processing of pri-miRNAs into pre-miRNAs.243 Recently, Wu et al reported that TAp63, a p53 family member, suppresses tumorigenesis and metastasis through the coordinate regulation of Dicer and miR-130b by directly binding and trans-activating the Dicer promoter, thereby demonstrating direct transcriptional regulation of Dicer by TAp63.241

3.5. Plasma cell free DNA

Circulating plasma cell free nucleic acids, now known to be mainly tumor derived,244, 245 were discovered by Mandel in 1948.246 They are detected in the blood of both healthy and sick individuals, and their concentration is generally related to tumor load and disease extent. Patients suffering from malignant diseases like EOC usually have increased amounts of circulating cell free nucleic acids. Tumors release genomic DNA into the systemic circulation, probably through cellular necrosis and apoptosis.247, 248 Such tumor-specific cell free DNA may be detected in the plasma by the presence of genetic and epigenetic alterations specific to the primary tumor.249251 This makes tumor-specific cell free DNA potential tumor biomarkers and therapeutic targets.

Fiegl and colleagues have found that persistence of RASSF1A DNA methylation of cell free DNA 1 year after primary surgery and adjuvant tamoxifen therapy is an independent predictor of poor outcome in breast cancer.252 In an orthotopic EOC mouse model, we have shown that cell free DNA levels correlate closely with tumor load and that those levels decline appreciably with chemotherapy.253 Consequently, cell-free DNA is a potential prognostic marker in several solid tumors, including ovarian cancer.254256 We have reported that cell-free DNA at concentrations >22,000 GE/mL is significantly associated with decreased patient survival. After adjusting for other clinical variables, preoperative cell-free DNA >22,000 GE/mL was an independent predictor for disease-specific survival. This suggests that serial monitoring of plasma nucleic acids may be useful for monitoring disease status and predict outcome. Zachariah and colleagues found higher levels of cell-free nuclear and mitochondrial DNA in ovarian cancer patients than in healthy controls but no correlation between cell-free DNA and prognosis.257 Further prospective studies on larger numbers of patients are needed to validate the prognostic value of cell-free DNA.

4. Combination of EOC prognostic biomarkers

The continuing discovery of new biomarkers of EOC raises the question of how to use them effectively. As already stated above, CA125 has often been combined with other biomarkers.26, 258260 Jacob et al have included it in their Risk of Malignancy Index (RMI), which combines serum CA125 level, ultrasound results, and menopausal status to help identify and triage high risk patients by estimating background cancer risk. They found that patients with RMI scores >200 had 42 times the background risk and that patients with lower scores had 0.15 times the risk.261

The development of effective biomarker combinations will ultimately rely on the proper design and appropriate use of systemic models and analyses.262, 263 One clinical trial in 200,000 women in the United Kingdom is now utilizing an algorithm to evaluate the ability of a 2-stage screening strategy to improve survival in ovarian cancer. More than 30 serum markers besides CA125 will be required to detect all patients in an initial phase of screening and will then need to be evaluated alone and in combination with CA125 by different investigators.25

5. Challenges and future directions

Despite years of research and mushrooming reports of new tumor biomarkers, the number of clinically useful markers is pitifully small.264 This is partly due to the complex nature of the tumor biomarkers themselves—especially those at the DNA, RNA, and protein levels—and partly due to the lack of acceptable standards for effectively evaluating and incorporating these newer biomarkers in the clinic. Thus, further validation and effective translation are needed. In addition, most prognostic studies in cancer are not protocol driven or prospective but retrospective.265 Retrospective studies offer the distinct advantage of large cohorts whose follow-up periods are long enough to allow assessment of a substantial number of outcomes of interest. Conversely, their disadvantages include unclear inclusion criteria, cohort completeness and follow-up; incomplete baseline data; lack of standardization of diagnostic and therapeutic procedures; and statistical analytical issues.3 Thus, validation and randomized clinical trials are needed to determine the true value of the new biomarkers for early diagnosis and improved survival and quality of life.

The first challenge is to establish acceptable criteria for evaluating new biomarkers and for standardizing the information that is gained in an integrated and clinically useful setting.266 In 2005, REMARK proposed guidelines meant to encourage more clarity and comprehensiveness in biomarker studies.267 It is imperative to establish integrated, clinically feasible criteria for tumor biomarker studies and to design and conduct prospective studies that use a reliable model, and enroll sufficient numbers of patients, and are properly and comprehensively analyzed statistically.268 Although it is important to seek a convergence of findings across multiple, independent data sets, it is only through the accumulation of evidence that tumor biomarkers in EOC can begin to be validated clinically.258, 259, 269

An important obstacle to the development of clinically useful tumor biomarkers is the availability of techniques for detecting them. Cancer-associated biomarkers in blood exist at low concentrations within complex mixtures of very abundant proteins, such as albumin and immunoglobulins, and there can be serious physiologic challenges to separating them out. Advances in nanotechnology are providing a completely new approach to this problem. For example, Liotta created a nanoparticle for biomarker “harvesting” that rapidly concentrates and amplifies scarce proteins for mass spectrometry analysis, multiple reaction monitoring, or immunoassay-based analysis.270

During the past decade, a growing knowledge of tumor biomarkers has been translated into targeted and personalized clinical therapies. One successful example is anti-angiogenesis therapy based on the developing knowledge of angiogenic biomarkers.28, 46, 48, 49, 50, 96 Another is the use of targeted drugs such as trastuzumab to target the HER2 proto-oncogene and EGFR inhibitors to block the EGFR signaling pathway.262, 263 In 2007, in an attempt to speed up the drug development process, improve safety and efficacy, and increase the cost efficiency of using tumor biomarkers to identify potentially useful new cancer drugs, the AACR-FDA-NCI Cancer Biomarkers Collaborative launched a national effort to clearly delineate the barriers to such research, develop recommendations for integrating biomarkers into the cancer drug development enterprise, and set in motion the necessary action plans and collaborations.271 Further efforts are needed to advance the use of biomarkers in cancer drug development and in the improvement of patient survival and quality of life.

In the near future, continued collaboration between clinicians, statisticians, and others should lead to establishment of clinically feasible criteria for biomarker studies, integration of reliable and statistically robust biomarker information into clinical treatment planning guidelines, and the development of more effective targeted strategies based on the knowledge gained from tumor biomarkers.

Acknowledgments

Portions of this work were supported by National Institutes of Health (CA 110793, 109298, P50 CA083639, P50 CA098258, CA128797, RC2GM092599, U54 CA151668), the Ovarian Cancer Research Fund, Inc. (Program Project Development Grant), the Department of Defense (OC073399, OC093146, BC085265), the Marcus Foundation, and the Betty Anne Asche Murray Distinguished Professorship, and NCI institutional Core Grant CA16672.

References

  • 1.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
  • 2.Ferte C, Andre F, Soria JC. Molecular circuits of solid tumors: prognostic and predictive tools for bedside use. Nat Rev Clin Oncol. 7:367–380. doi: 10.1038/nrclinonc.2010.84. [DOI] [PubMed] [Google Scholar]
  • 3.Altman DG, Riley RD. Primer: an evidence-based approach to prognostic markers. Nat Clin Pract Oncol. 2005;2:466–472. doi: 10.1038/ncponc0287. [DOI] [PubMed] [Google Scholar]
  • 4.Jacobs I, Bast RC., Jr The CA 125 tumour-associated antigen: a review of the literature. Hum Reprod. 1989;4:1–12. doi: 10.1093/oxfordjournals.humrep.a136832. [DOI] [PubMed] [Google Scholar]
  • 5.Bast RC, Jr, Feeney M, Lazarus H, Nadler LM, Colvin RB, Knapp RC. Reactivity of a monoclonal antibody with human ovarian carcinoma. J Clin Invest. 1981;68:1331–1337. doi: 10.1172/JCI110380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Einhorn N, Bast RC, Jr, Knapp RC, Tjernberg B, Zurawski VR., Jr Preoperative evaluation of serum CA 125 levels in patients with primary epithelial ovarian cancer. Obstet Gynecol. 1986;67:414–416. [PubMed] [Google Scholar]
  • 7.Vasilev SA, Schlaerth JB, Campeau J, Morrow CP. Serum CA 125 levels in preoperative evaluation of pelvic masses. Obstet Gynecol. 1988;71:751–756. [PubMed] [Google Scholar]
  • 8.Yarbro JW, Page DL, Fielding LP, Partridge EE, Murphy GP. American Joint Committee on Cancer prognostic factors consensus conference. Cancer. 1999;86:2436–2446. doi: 10.1002/(sici)1097-0142(19991201)86:11<2436::aid-cncr35>3.0.co;2-#. [DOI] [PubMed] [Google Scholar]
  • 9.Fayers PM, Rustin G, Wood R, et al. The prognostic value of serum CA 125 in patients with advanced ovarian carcinoma: an analysis of 573 patients by the Medical Research Council Working Party on Gynaecological Cancer. Int J Gynecol Cancer. 1993;3:285–292. doi: 10.1046/j.1525-1438.1993.03050285.x. [DOI] [PubMed] [Google Scholar]
  • 10.Rustin GJ, Bast RC, Jr, Kelloff GJ, et al. Use of CA-125 in clinical trial evaluation of new therapeutic drugs for ovarian cancer. Clin Cancer Res. 2004;10:3919–3926. doi: 10.1158/1078-0432.CCR-03-0787. [DOI] [PubMed] [Google Scholar]
  • 11.Anderson GL. Ovarian cancer biomarker screening: still too early to tell. Womens Health (Lond Engl) 6:487–490. doi: 10.2217/whe.10.33. [DOI] [PubMed] [Google Scholar]
  • 12.Seidman JD, Yemelyanova AV, Khedmati F, et al. Prognostic factors for stage I ovarian carcinoma. Int J Gynecol Pathol. 29:1–7. doi: 10.1097/PGP.0b013e3181af2372. [DOI] [PubMed] [Google Scholar]
  • 13.Kaesemann H, Caffier H, Hoffmann FJ, et al. [Monoclonal antibodies in the diagnosis and follow-up of ovarian cancer. CA 125 as a tumor marker. A cooperative study of the Gynecologic Tumor Marker Group (GTMG)] Klin Wochenschr. 1986;64:781–785. doi: 10.1007/BF01732187. [DOI] [PubMed] [Google Scholar]
  • 14.Makar AP, Kristensen GB, Kaern J, Bormer OP, Abeler VM, Trope CG. Prognostic value of pre- and postoperative serum CA 125 levels in ovarian cancer: new aspects and multivariate analysis. Obstet Gynecol. 1992;79:1002–1010. [PubMed] [Google Scholar]
  • 15.Han LY, Karavasilis V, Hagen T, et al. Doubling time of serum CA125 is an independent prognostic factor for survival in patients with ovarian cancer relapsing after first-line chemotherapy. Eur J Cancer. 46:1359–1364. doi: 10.1016/j.ejca.2010.02.012. [DOI] [PubMed] [Google Scholar]
  • 16.Gronlund B, Hogdall C, Hilden J, Engelholm SA, Hogdall EV, Hansen HH. Should CA-125 response criteria be preferred to response evaluation criteria in solid tumors (RECIST) for prognostication during second-line chemotherapy of ovarian carcinoma? J Clin Oncol. 2004;22:4051–4058. doi: 10.1200/JCO.2004.10.028. [DOI] [PubMed] [Google Scholar]
  • 17.Gadducci A, Cosio S, Tana R, Genazzani AR. Serum and tissue biomarkers as predictive and prognostic variables in epithelial ovarian cancer. Crit Rev Oncol Hematol. 2009;69:12–27. doi: 10.1016/j.critrevonc.2008.05.001. [DOI] [PubMed] [Google Scholar]
  • 18.Crawford SM, Peace J. Does the nadir CA125 concentration predict a long-term outcome after chemotherapy for carcinoma of the ovary? Ann Oncol. 2005;16:47–50. doi: 10.1093/annonc/mdi012. [DOI] [PubMed] [Google Scholar]
  • 19.Gadducci A, Cosio S, Fanucchi A, Negri S, Cristofani R, Genazzani AR. The predictive and prognostic value of serum CA 125 half-life during paclitaxel/platinum-based chemotherapy in patients with advanced ovarian carcinoma. Gynecol Oncol. 2004;93:131–136. doi: 10.1016/j.ygyno.2003.12.043. [DOI] [PubMed] [Google Scholar]
  • 20.Meyer T, Rustin GJ. Role of tumour markers in monitoring epithelial ovarian cancer. Br J Cancer. 2000;82:1535–1538. doi: 10.1054/bjoc.2000.1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Rocconi RP, Matthews KS, Kemper MK, Hoskins KE, Huh WK, Straughn JM., Jr The timing of normalization of CA-125 levels during primary chemotherapy is predictive of survival in patients with epithelial ovarian cancer. Gynecol Oncol. 2009;114:242–245. doi: 10.1016/j.ygyno.2009.04.019. [DOI] [PubMed] [Google Scholar]
  • 22.Rustin GJ, Nelstrop AE, Bentzen SM, Bond SJ, McClean P. Selection of active drugs for ovarian cancer based on CA-125 and standard response rates in phase II trials. J Clin Oncol. 2000;18:1733–1739. doi: 10.1200/JCO.2000.18.8.1733. [DOI] [PubMed] [Google Scholar]
  • 23.Piccart MJ, Gore M, Ten Bokkel Huinink W, et al. Docetaxel: an active new drug for treatment of advanced epithelial ovarian cancer. J Natl Cancer Inst. 1995;87:676–681. doi: 10.1093/jnci/87.9.676. [DOI] [PubMed] [Google Scholar]
  • 24.Karam AK, Karlan BY. Ovarian cancer: the duplicity of CA125 measurement. Nat Rev Clin Oncol. 7:335–339. doi: 10.1038/nrclinonc.2010.44. [DOI] [PubMed] [Google Scholar]
  • 25.Bast RC, Jr, Badgwell D, Lu Z, et al. New tumor markers: CA125 and beyond. Int J Gynecol Cancer. 2005;15(Suppl 3):274–281. doi: 10.1111/j.1525-1438.2005.00441.x. [DOI] [PubMed] [Google Scholar]
  • 26.Menon U, Gentry-Maharaj A, Hallett R, et al. Sensitivity and specificity of multimodal and ultrasound screening for ovarian cancer, and stage distribution of detected cancers: results of the prevalence screen of the UK Collaborative Trial of Ovarian Cancer Screening (UKCTOCS) Lancet Oncol. 2009;10:327–340. doi: 10.1016/S1470-2045(09)70026-9. [DOI] [PubMed] [Google Scholar]
  • 27.Coticchia CM, Yang J, Moses MA. Ovarian cancer biomarkers: current options and future promise. J Natl Compr Canc Netw. 2008;6:795–802. doi: 10.6004/jnccn.2008.0059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Thaker PH, Deavers M, Celestino J, et al. EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin Cancer Res. 2004;10:5145–5150. doi: 10.1158/1078-0432.CCR-03-0589. [DOI] [PubMed] [Google Scholar]
  • 29.Paley PJ, Staskus KA, Gebhard K, et al. Vascular endothelial growth factor expression in early stage ovarian carcinoma. Cancer. 1997;80:98–106. doi: 10.1002/(sici)1097-0142(19970701)80:1<98::aid-cncr13>3.0.co;2-a. [DOI] [PubMed] [Google Scholar]
  • 30.Siddiqui GK, Elmasry K, Wong Te Fong AC, et al. Prognostic significance of intratumoral vascular endothelial growth factor as a marker of tumour angiogenesis in epithelial ovarian cancer. Eur J Gynaecol Oncol. 31:156–159. [PubMed] [Google Scholar]
  • 31.Sonmezer M, Gungor M, Ensari A, Ortac F. Prognostic significance of tumor angiogenesis in epithelial ovarian cancer: in association with transforming growth factor beta and vascular endothelial growth factor. Int J Gynecol Cancer. 2004;14:82–88. doi: 10.1111/j.1048-891x.2004.14202.x. [DOI] [PubMed] [Google Scholar]
  • 32.Chen CA, Cheng WF, Lee CN, et al. Serum vascular endothelial growth factor in epithelial ovarian neoplasms: correlation with patient survival. Gynecol Oncol. 1999;74:235–240. doi: 10.1006/gyno.1999.5418. [DOI] [PubMed] [Google Scholar]
  • 33.Spannuth WA, Nick AM, Jennings NB, et al. Functional significance of VEGFR-2 on ovarian cancer cells. Int J Cancer. 2009;124:1045–1053. doi: 10.1002/ijc.24028. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bamberger ES, Perrett CW. Angiogenesis in epithelian ovarian cancer. Mol Pathol. 2002;55:348–359. doi: 10.1136/mp.55.6.348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tempfer C, Obermair A, Hefler L, Haeusler G, Gitsch G, Kainz C. Vascular endothelial growth factor serum concentrations in ovarian cancer. Obstet Gynecol. 1998;92:360–363. doi: 10.1016/s0029-7844(98)00190-2. [DOI] [PubMed] [Google Scholar]
  • 36.Folkman J. Endothelial cells and angiogenic growth factors in cancer growth and metastasis. Introduction. Cancer Metastasis Rev. 1990;9:171–174. doi: 10.1007/BF00046358. [DOI] [PubMed] [Google Scholar]
  • 37.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
  • 38.Yokoyama Y, Charnock-Jones DS, Licence D, et al. Vascular endothelial growth factor-D is an independent prognostic factor in epithelial ovarian carcinoma. Br J Cancer. 2003;88:237–244. doi: 10.1038/sj.bjc.6600701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Engels K, du Bois A, Harter P, et al. VEGF-A and i-NOS expression are prognostic factors in serous epithelial ovarian carcinomas after complete surgical resection. J Clin Pathol. 2009;62:448–454. doi: 10.1136/jcp.2008.063859. [DOI] [PubMed] [Google Scholar]
  • 40.Halder J, Lin YG, Merritt WM, et al. Therapeutic efficacy of a novel focal adhesion kinase inhibitor TAE226 in ovarian carcinoma. Cancer Res. 2007;67:10976–10983. doi: 10.1158/0008-5472.CAN-07-2667. [DOI] [PubMed] [Google Scholar]
  • 41.Charlesworth PJ, Harris AL. Mechanisms of disease: angiogenesis in urologic malignancies. Nat Clin Pract Urol. 2006;3:157–169. doi: 10.1038/ncpuro0434. [DOI] [PubMed] [Google Scholar]
  • 42.Sood AK, Fletcher MS, Zahn CM, et al. The clinical significance of tumor cell-lined vasculature in ovarian carcinoma: implications for anti-vasculogenic therapy. Cancer Biol Ther. 2002;1:661–664. doi: 10.4161/cbt.316. [DOI] [PubMed] [Google Scholar]
  • 43.Gerber HP, McMurtrey A, Kowalski J, et al. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3'-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation. J Biol Chem. 1998;273:30336–30343. doi: 10.1074/jbc.273.46.30336. [DOI] [PubMed] [Google Scholar]
  • 44.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–2342. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 45.Jain RK, Duda DG, Clark JW, Loeffler JS. Lessons from phase III clinical trials on anti-VEGF therapy for cancer. Nat Clin Pract Oncol. 2006;3:24–40. doi: 10.1038/ncponc0403. [DOI] [PubMed] [Google Scholar]
  • 46.Landen CN, Kinch MS, Sood AK. EphA2 as a target for ovarian cancer therapy. Expert Opin Ther Targets. 2005;9:1179–1187. doi: 10.1517/14728222.9.6.1179. [DOI] [PubMed] [Google Scholar]
  • 47.Lin YG, Han LY, Kamat AA, et al. EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer. 2007;109:332–340. doi: 10.1002/cncr.22415. [DOI] [PubMed] [Google Scholar]
  • 48.Chon HS, Hu W, Kavanagh JJ. Targeted therapies in gynecologic cancers. Curr Cancer Drug Targets. 2006;6:333–363. doi: 10.2174/156800906777441799. [DOI] [PubMed] [Google Scholar]
  • 49.Landen CN, Jr, Lu C, Han LY, et al. Efficacy and antivascular effects of EphA2 reduction with an agonistic antibody in ovarian cancer. J Natl Cancer Inst. 2006;98:1558–1570. doi: 10.1093/jnci/djj414. [DOI] [PubMed] [Google Scholar]
  • 50.Yang N, Kaur S, Volinia S, et al. MicroRNA microarray identifies Let-7i as a novel biomarker and therapeutic target in human epithelial ovarian cancer. Cancer Res. 2008;68:10307–10314. doi: 10.1158/0008-5472.CAN-08-1954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lu C, Han HD, Mangala LS, et al. Regulation of tumor angiogenesis by EZH2. Cancer Cell. 18:185–197. doi: 10.1016/j.ccr.2010.06.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Merritt WM, Sood AK. Markers of angiogenesis in ovarian cancer. Dis Markers. 2007;23:419–431. doi: 10.1155/2007/257602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Landen CN, Jr, Chavez-Reyes A, Bucana C, et al. Therapeutic EphA2 gene targeting in vivo using neutral liposomal small interfering RNA delivery. Cancer Res. 2005;65:6910–6918. doi: 10.1158/0008-5472.CAN-05-0530. [DOI] [PubMed] [Google Scholar]
  • 54.Berek JS, Chung C, Kaldi K, Watson JM, Knox RM, Martinez-Maza O. Serum interleukin-6 levels correlate with disease status in patients with epithelial ovarian cancer. Am J Obstet Gynecol. 1991;164:1038–1042. doi: 10.1016/0002-9378(91)90582-c. discussion 1042-1033. [DOI] [PubMed] [Google Scholar]
  • 55.Scambia G, Testa U, Benedetti Panici P, et al. Prognostic significance of interleukin 6 serum levels in patients with ovarian cancer. Br J Cancer. 1995;71:354–356. doi: 10.1038/bjc.1995.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Tempfer C, Zeisler H, Sliutz G, Haeusler G, Hanzal E, Kainz C. Serum evaluation of interleukin 6 in ovarian cancer patients. Gynecol Oncol. 1997;66:27–30. doi: 10.1006/gyno.1997.4726. [DOI] [PubMed] [Google Scholar]
  • 57.Plante M, Rubin SC, Wong GY, Federici MG, Finstad CL, Gastl GA. Interleukin-6 level in serum and ascites as a prognostic factor in patients with epithelial ovarian cancer. Cancer. 1994;73:1882–1888. doi: 10.1002/1097-0142(19940401)73:7<1882::aid-cncr2820730718>3.0.co;2-r. [DOI] [PubMed] [Google Scholar]
  • 58.Gorelik E, Landsittel DP, Marrangoni AM, et al. Multiplexed immunobead-based cytokine profiling for early detection of ovarian cancer. Cancer Epidemiol Biomarkers Prev. 2005;14:981–987. doi: 10.1158/1055-9965.EPI-04-0404. [DOI] [PubMed] [Google Scholar]
  • 59.Nilsson MB, Langley RR, Fidler IJ. Interleukin-6, secreted by human ovarian carcinoma cells, is a potent proangiogenic cytokine. Cancer Res. 2005;65:10794–10800. doi: 10.1158/0008-5472.CAN-05-0623. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lokshin AE, Winans M, Landsittel D, et al. Circulating IL-8 and anti-IL-8 autoantibody in patients with ovarian cancer. Gynecol Oncol. 2006;102:244–251. doi: 10.1016/j.ygyno.2005.12.011. [DOI] [PubMed] [Google Scholar]
  • 61.Xie K. Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev. 2001;12:375–391. doi: 10.1016/s1359-6101(01)00016-8. [DOI] [PubMed] [Google Scholar]
  • 62.Kassim SK, El-Salahy EM, Fayed ST, et al. Vascular endothelial growth factor and interleukin-8 are associated with poor prognosis in epithelial ovarian cancer patients. Clin Biochem. 2004;37:363–369. doi: 10.1016/j.clinbiochem.2004.01.014. [DOI] [PubMed] [Google Scholar]
  • 63.Koch AE, Polverini PJ, Kunkel SL, et al. Interleukin-8 as a macrophage-derived mediator of angiogenesis. Science. 1992;258:1798–1801. doi: 10.1126/science.1281554. [DOI] [PubMed] [Google Scholar]
  • 64.Merritt WM, Lin YG, Spannuth WA, et al. Effect of interleukin-8 gene silencing with liposome-encapsulated small interfering RNA on ovarian cancer cell growth. J Natl Cancer Inst. 2008;100:359–372. doi: 10.1093/jnci/djn024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Uslu R, Sanli UA, Dikmen Y, et al. Predictive value of serum interleukin-8 levels in ovarian cancer patients treated with paclitaxel-containing regimens. Int J Gynecol Cancer. 2005;15:240–245. doi: 10.1111/j.1525-1438.2005.15210.x. [DOI] [PubMed] [Google Scholar]
  • 66.Balch CM, Gershenwald JE, Soong SJ, et al. Final version of 2009 AJCC melanoma staging and classification. J Clin Oncol. 2009;27:6199–6206. doi: 10.1200/JCO.2009.23.4799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Miyoshi J, Takai Y. Molecular perspective on tight-junction assembly and epithelial polarity. Adv Drug Deliv Rev. 2005;57:815–855. doi: 10.1016/j.addr.2005.01.008. [DOI] [PubMed] [Google Scholar]
  • 68.Morin PJ. Claudin proteins in human cancer: promising new targets for diagnosis and therapy. Cancer Res. 2005;65:9603–9606. doi: 10.1158/0008-5472.CAN-05-2782. [DOI] [PubMed] [Google Scholar]
  • 69.Tassi RA, Bignotti E, Falchetti M, et al. Claudin-7 expression in human epithelial ovarian cancer. Int J Gynecol Cancer. 2008;18:1262–1271. doi: 10.1111/j.1525-1438.2008.01194.x. [DOI] [PubMed] [Google Scholar]
  • 70.Huang YH, Bao Y, Peng W, et al. Claudin-3 gene silencing with siRNA suppresses ovarian tumor growth and metastasis. Proc Natl Acad Sci U S A. 2009;106:3426–3430. doi: 10.1073/pnas.0813348106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Choi YL, Kim J, Kwon MJ, et al. Expression profile of tight junction protein claudin 3 and claudin 4 in ovarian serous adenocarcinoma with prognostic correlation. Histol Histopathol. 2007;22:1185–1195. doi: 10.14670/HH-22.1185. [DOI] [PubMed] [Google Scholar]
  • 72.Agarwal R, D'Souza T, Morin PJ. Claudin-3 and claudin-4 expression in ovarian epithelial cells enhances invasion and is associated with increased matrix metalloproteinase-2 activity. Cancer Res. 2005;65:7378–7385. doi: 10.1158/0008-5472.CAN-05-1036. [DOI] [PubMed] [Google Scholar]
  • 73.Yurchenco PD, Schittny JC. Molecular architecture of basement membranes. FASEB J. 1990;4:1577–1590. doi: 10.1096/fasebj.4.6.2180767. [DOI] [PubMed] [Google Scholar]
  • 74.Ellerbroek SM, Fishman DA, Kearns AS, Bafetti LM, Stack MS. Ovarian carcinoma regulation of matrix metalloproteinase-2 and membrane type 1 matrix metalloproteinase through beta1 integrin. Cancer Res. 1999;59:1635–1641. [PubMed] [Google Scholar]
  • 75.Fishman DA, Bafetti LM, Stack MS. Membrane-type matrix metalloproteinase expression and matrix metalloproteinase-2 activation in primary human ovarian epithelial carcinoma cells. Invasion Metastasis. 1996;16:150–159. [PubMed] [Google Scholar]
  • 76.Davidson B, Goldberg I, Gotlieb WH, et al. High levels of MMP-2, MMP-9, MT1-MMP and TIMP-2 mRNA correlate with poor survival in ovarian carcinoma. Clin Exp Metastasis. 1999;17:799–808. doi: 10.1023/a:1006723011835. [DOI] [PubMed] [Google Scholar]
  • 77.Stack MS, Ellerbroek SM, Fishman DA. The role of proteolytic enzymes in the pathology of epithelial ovarian carcinoma. Int J Oncol. 1998;12:569–576. doi: 10.3892/ijo.12.3.569. [DOI] [PubMed] [Google Scholar]
  • 78.Kamat AA, Fletcher M, Gruman LM, et al. The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res. 2006;12:1707–1714. doi: 10.1158/1078-0432.CCR-05-2338. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Stadlmann S, Pollheimer J, Moser PL, et al. Cytokine-regulated expression of collagenase-2 (MMP-8) is involved in the progression of ovarian cancer. Eur J Cancer. 2003;39:2499–2505. doi: 10.1016/j.ejca.2003.08.011. [DOI] [PubMed] [Google Scholar]
  • 80.Torng PL, Mao TL, Chan WY, Huang SC, Lin CT. Prognostic significance of stromal metalloproteinase-2 in ovarian adenocarcinoma and its relation to carcinoma progression. Gynecol Oncol. 2004;92:559–567. doi: 10.1016/j.ygyno.2003.11.011. [DOI] [PubMed] [Google Scholar]
  • 81.Davidson B, Goldberg I, Gotlieb WH, et al. The prognostic value of metalloproteinases and angiogenic factors in ovarian carcinoma. Mol Cell Endocrinol. 2002;187:39–45. doi: 10.1016/s0303-7207(01)00709-2. [DOI] [PubMed] [Google Scholar]
  • 82.Ozalp S, Tanir HM, Yalcin OT, Kabukcuoglu S, Oner U, Uray M. Prognostic value of matrix metalloproteinase-9 (gelatinase-B) expression in epithelial ovarian tumors. Eur J Gynaecol Oncol. 2003;24:417–420. [PubMed] [Google Scholar]
  • 83.Westerlund A, Apaja-Sarkkinen M, Hoyhtya M, Puistola U, Turpeenniemi-Hujanen T. Gelatinase A-immunoreactive protein in ovarian lesions- prognostic value in epithelial ovarian cancer. Gynecol Oncol. 1999;75:91–98. doi: 10.1006/gyno.1999.5533. [DOI] [PubMed] [Google Scholar]
  • 84.Schaller MD, Parsons JT. Focal adhesion kinase: an integrin-linked protein tyrosine kinase. Trends Cell Biol. 1993;3:258–262. doi: 10.1016/0962-8924(93)90053-4. [DOI] [PubMed] [Google Scholar]
  • 85.Owens LV, Xu L, Craven RJ, et al. Overexpression of the focal adhesion kinase (p125FAK) in invasive human tumors. Cancer Res. 1995;55:2752–2755. [PubMed] [Google Scholar]
  • 86.Cance WG, Harris JE, Iacocca MV, et al. Immunohistochemical analyses of focal adhesion kinase expression in benign and malignant human breast and colon tissues: correlation with preinvasive and invasive phenotypes. Clin Cancer Res. 2000;6:2417–2423. [PubMed] [Google Scholar]
  • 87.Owens LV, Xu L, Dent GA, et al. Focal adhesion kinase as a marker of invasive potential in differentiated human thyroid cancer. Ann Surg Oncol. 1996;3:100–105. doi: 10.1007/BF02409059. [DOI] [PubMed] [Google Scholar]
  • 88.Ayaki M, Komatsu K, Mukai M, et al. Reduced expression of focal adhesion kinase in liver metastases compared with matched primary human colorectal adenocarcinomas. Clin Cancer Res. 2001;7:3106–3112. [PubMed] [Google Scholar]
  • 89.Judson PL, He X, Cance WG, Van Le L. Overexpression of focal adhesion kinase, a protein tyrosine kinase, in ovarian carcinoma. Cancer. 1999;86:1551–1556. doi: 10.1002/(sici)1097-0142(19991015)86:6<1551::aid-cncr23>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
  • 90.Sood AK, Coffin JE, Schneider GB, et al. Biological significance of focal adhesion kinase in ovarian cancer: role in migration and invasion. Am J Pathol. 2004;165:1087–1095. doi: 10.1016/S0002-9440(10)63370-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Shahzad MM, Lu C, Lee JW, et al. Dual targeting of EphA2 and FAK in ovarian carcinoma. Cancer Biol Ther. 2009;8:1027–1034. doi: 10.4161/cbt.8.11.8523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Labiche A, Heutte N, Herlin P, Chasle J, Gauduchon P, Elie N. Stromal compartment as a survival prognostic factor in advanced ovarian carcinoma. Int J Gynecol Cancer. 20:28–33. doi: 10.1111/IGC.0b013e3181bda1cb. [DOI] [PubMed] [Google Scholar]
  • 93.Weidner N. Tumor angiogenesis: review of current applications in tumor prognostication. Semin Diagn Pathol. 1993;10:302–313. [PubMed] [Google Scholar]
  • 94.Thijssen VL, Brandwijk RJ, Dings RP, Griffioen AW. Angiogenesis gene expression profiling in xenograft models to study cellular interactions. Exp Cell Res. 2004;299:286–293. doi: 10.1016/j.yexcr.2004.06.014. [DOI] [PubMed] [Google Scholar]
  • 95.Mendiola M, Barriuso J, Redondo A, et al. Angiogenesis-related gene expression profile with independent prognostic value in advanced ovarian carcinoma. PLoS One. 2008;3:e4051. doi: 10.1371/journal.pone.0004051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Sessa C, Guibal A, Del Conte G, Ruegg C. Biomarkers of angiogenesis for the development of antiangiogenic therapies in oncology: tools or decorations? Nat Clin Pract Oncol. 2008;5:378–391. doi: 10.1038/ncponc1150. [DOI] [PubMed] [Google Scholar]
  • 97.Gadducci A, Viacava P, Cosio S, et al. Vascular endothelial growth factor (VEGF) expression in primary tumors and peritoneal metastases from patients with advanced ovarian carcinoma. Anticancer Res. 2003;23:3001–3008. [PubMed] [Google Scholar]
  • 98.Yamamoto S, Konishi I, Mandai M, et al. Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. Br J Cancer. 1997;76:1221–1227. doi: 10.1038/bjc.1997.537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Grunwald V, Hidalgo M. The epidermal growth factor receptor: a new target for anticancer therapy. Curr Probl Cancer. 2002;26:109–164. doi: 10.1067/mcn.2002.125874. [DOI] [PubMed] [Google Scholar]
  • 100.See HT, Kavanagh JJ, Hu W, Bast RC. Targeted therapy for epithelial ovarian cancer: current status and future prospects. Int J Gynecol Cancer. 2003;13:701–734. doi: 10.1111/j.1525-1438.2003.13601.x. [DOI] [PubMed] [Google Scholar]
  • 101.Mass RD. The HER receptor family: a rich target for therapeutic development. Int J Radiat Oncol Biol Phys. 2004;58:932–940. doi: 10.1016/j.ijrobp.2003.09.093. [DOI] [PubMed] [Google Scholar]
  • 102.Hsieh AC, Moasser MM. Targeting HER proteins in cancer therapy and the role of the non-target HER3. Br J Cancer. 2007;97:453–457. doi: 10.1038/sj.bjc.6603910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Vaidya AP, Parnes AD, Seiden MV. Rationale and clinical experience with epidermal growth factor receptor inhibitors in gynecologic malignancies. Curr Treat Options Oncol. 2005;6:103–114. doi: 10.1007/s11864-005-0018-x. [DOI] [PubMed] [Google Scholar]
  • 104.Salomon DS, Brandt R, Ciardiello F, Normanno N. Epidermal growth factor-related peptides and their receptors in human malignancies. Crit Rev Oncol Hematol. 1995;19:183–232. doi: 10.1016/1040-8428(94)00144-i. [DOI] [PubMed] [Google Scholar]
  • 105.Maurizi M, Almadori G, Ferrandina G, et al. Prognostic significance of epidermal growth factor receptor in laryngeal squamous cell carcinoma. Br J Cancer. 1996;74:1253–1257. doi: 10.1038/bjc.1996.525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Fischer-Colbrie J, Witt A, Heinzl H, et al. EGFR and steroid receptors in ovarian carcinoma: comparison with prognostic parameters and outcome of patients. Anticancer Res. 1997;17:613–619. [PubMed] [Google Scholar]
  • 107.Kersemaekers AM, Fleuren GJ, Kenter GG, et al. Oncogene alterations in carcinomas of the uterine cervix: overexpression of the epidermal growth factor receptor is associated with poor prognosis. Clin Cancer Res. 1999;5:577–586. [PubMed] [Google Scholar]
  • 108.Mellon K, Wright C, Kelly P, Horne CH, Neal DE. Long-term outcome related to epidermal growth factor receptor status in bladder cancer. J Urol. 1995;153:919–925. [PubMed] [Google Scholar]
  • 109.Nicholson RI, Gee JM, Harper ME. EGFR and cancer prognosis. Eur J Cancer. 2001;37(Suppl 4):S9–S15. doi: 10.1016/s0959-8049(01)00231-3. [DOI] [PubMed] [Google Scholar]
  • 110.Maihle NJ, Baron AT, Barrette BA, et al. EGF/ErbB receptor family in ovarian cancer. Cancer Treat Res. 2002;107:247–258. doi: 10.1007/978-1-4757-3587-1_11. [DOI] [PubMed] [Google Scholar]
  • 111.Tanaka Y, Terai Y, Tanabe A, et al. Prognostic effect of epidermal growth factor receptor gene mutations and the aberrant phosphorylation of Akt and ERK in ovarian cancer. Cancer Biol Ther. 11 doi: 10.4161/cbt.11.1.13877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Xia W, Wei Y, Du Y, et al. Nuclear expression of epidermal growth factor receptor is a novel prognostic value in patients with ovarian cancer. Mol Carcinog. 2009;48:610–617. doi: 10.1002/mc.20504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hynes NE. Amplification and overexpression of the erbB-2 gene in human tumors: its involvement in tumor development, significance as a prognostic factor, and potential as a target for cancer therapy. Semin Cancer Biol. 1993;4:19–26. [PubMed] [Google Scholar]
  • 114.Serrano-Olvera A, Duenas-Gonzalez A, Gallardo-Rincon D, Candelaria M, De la Garza-Salazar J. Prognostic, predictive and therapeutic implications of HER2 in invasive epithelial ovarian cancer. Cancer Treat Rev. 2006;32:180–190. doi: 10.1016/j.ctrv.2006.01.001. [DOI] [PubMed] [Google Scholar]
  • 115.Slamon DJ, Godolphin W, Jones LA, et al. Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancer. Science. 1989;244:707–712. doi: 10.1126/science.2470152. [DOI] [PubMed] [Google Scholar]
  • 116.Meden H, Marx D, Roegglen T, Schauer A, Kuhn W. Overexpression of the oncogene c-erbB-2 (HER2/neu) and response to chemotherapy in patients with ovarian cancer. Int J Gynecol Pathol. 1998;17:61–65. doi: 10.1097/00004347-199801000-00011. [DOI] [PubMed] [Google Scholar]
  • 117.Tanner B, Kreutz E, Weikel W, et al. Prognostic significance of c-erB-2 mRNA in ovarian carcinoma. Gynecol Oncol. 1996;62:268–277. doi: 10.1006/gyno.1996.0226. [DOI] [PubMed] [Google Scholar]
  • 118.Felip E, Del Campo JM, Rubio D, Vidal MT, Colomer R, Bermejo B. Overexpression of c-erbB-2 in epithelial ovarian cancer. Prognostic value and relationship with response to chemotherapy. Cancer. 1995;75:2147–2152. doi: 10.1002/1097-0142(19950415)75:8<2147::aid-cncr2820750818>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
  • 119.Hogdall EV, Christensen L, Kjaer SK, et al. Distribution of HER-2 overexpression in ovarian carcinoma tissue and its prognostic value in patients with ovarian carcinoma: from the Danish MALOVA Ovarian Cancer Study. Cancer. 2003;98:66–73. doi: 10.1002/cncr.11476. [DOI] [PubMed] [Google Scholar]
  • 120.Camilleri-Broet S, Hardy-Bessard AC, Le Tourneau A, et al. HER-2 overexpression is an independent marker of poor prognosis of advanced primary ovarian carcinoma: a multicenter study of the GINECO group. Ann Oncol. 2004;15:104–112. doi: 10.1093/annonc/mdh021. [DOI] [PubMed] [Google Scholar]
  • 121.Gordon MS, Matei D, Aghajanian C, et al. Clinical activity of pertuzumab (rhuMAb 2C4), a HER dimerization inhibitor, in advanced ovarian cancer: potential predictive relationship with tumor HER2 activation status. J Clin Oncol. 2006;24:4324–4332. doi: 10.1200/JCO.2005.05.4221. [DOI] [PubMed] [Google Scholar]
  • 122.Yarden Y, Sliwkowski MX. Untangling the ErbB signalling network. Nat Rev Mol Cell Biol. 2001;2:127–137. doi: 10.1038/35052073. [DOI] [PubMed] [Google Scholar]
  • 123.Rowinsky EK. The erbB family: targets for therapeutic development against cancer and therapeutic strategies using monoclonal antibodies and tyrosine kinase inhibitors. Annu Rev Med. 2004;55:433–457. doi: 10.1146/annurev.med.55.091902.104433. [DOI] [PubMed] [Google Scholar]
  • 124.Amler LC. HER3 mRNA as a predictive biomarker in anticancer therapy. Expert Opin Biol Ther. 10:1343–1355. doi: 10.1517/14712598.2010.512003. [DOI] [PubMed] [Google Scholar]
  • 125.Junttila TT, Akita RW, Parsons K, et al. Ligand-independent HER2/HER3/PI3K complex is disrupted by trastuzumab and is effectively inhibited by the PI3K inhibitor GDC-0941. Cancer Cell. 2009;15:429–440. doi: 10.1016/j.ccr.2009.03.020. [DOI] [PubMed] [Google Scholar]
  • 126.Tanner B, Hasenclever D, Stern K, et al. ErbB-3 predicts survival in ovarian cancer. J Clin Oncol. 2006;24:4317–4323. doi: 10.1200/JCO.2005.04.8397. [DOI] [PubMed] [Google Scholar]
  • 127.Tzahar E, Waterman H, Chen X, et al. A hierarchical network of interreceptor interactions determines signal transduction by Neu differentiation factor/neuregulin and epidermal growth factor. Mol Cell Biol. 1996;16:5276–5287. doi: 10.1128/mcb.16.10.5276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Naresh A, Long W, Vidal GA, et al. The ERBB4/HER4 intracellular domain 4ICD is a BH3-only protein promoting apoptosis of breast cancer cells. Cancer Res. 2006;66:6412–6420. doi: 10.1158/0008-5472.CAN-05-2368. [DOI] [PubMed] [Google Scholar]
  • 129.de Graeff P, Crijns AP, de Jong S, et al. Modest effect of p53, EGFR and HER-2/neu on prognosis in epithelial ovarian cancer: a meta-analysis. Br J Cancer. 2009;101:149–159. doi: 10.1038/sj.bjc.6605112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Baron AT, Wilken JA, Haggstrom DE, Goodrich ST, Maihle NJ. Clinical implementation of soluble EGFR (sEGFR) as a theragnostic serum biomarker of breastm lung and ovarian cancer. IDrugs. 2009;12:302–308. [PubMed] [Google Scholar]
  • 131.Palayekar MJ, Herzog TJ. The emerging role of epidermal growth factor receptor inhibitors in ovarian cancer. Int J Gynecol Cancer. 2008;18:879–890. doi: 10.1111/j.1525-1438.2007.01144.x. [DOI] [PubMed] [Google Scholar]
  • 132.Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody, trastuzumab, in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J Clin Oncol. 2003;21:283–290. doi: 10.1200/JCO.2003.10.104. [DOI] [PubMed] [Google Scholar]
  • 133.Frederick PJ, Straughn JM, Jr, Alvarez RD, Buchsbaum DJ. Preclinical studies and clinical utilization of monoclonal antibodies in epithelial ovarian cancer. Gynecol Oncol. 2009;113:384–390. doi: 10.1016/j.ygyno.2009.01.008. [DOI] [PubMed] [Google Scholar]
  • 134.Greenblatt MS, Bennett WP, Hollstein M, Harris CC. Mutations in the p53 tumor suppressor gene: clues to cancer etiology and molecular pathogenesis. Cancer Res. 1994;54:4855–4878. [PubMed] [Google Scholar]
  • 135.Berchuck A, Kohler MF, Marks JR, Wiseman R, Boyd J, Bast RC., Jr The p53 tumor suppressor gene frequently is altered in gynecologic cancers. Am J Obstet Gynecol. 1994;170:246–252. doi: 10.1016/s0002-9378(94)70414-7. [DOI] [PubMed] [Google Scholar]
  • 136.Kastan MB, Onyekwere O, Sidransky D, Vogelstein B, Craig RW. Participation of p53 protein in the cellular response to DNA damage. Cancer Res. 1991;51:6304–6311. [PubMed] [Google Scholar]
  • 137.Fritsche M, Haessler C, Brandner G. Induction of nuclear accumulation of the tumor-suppressor protein p53 by DNA-damaging agents. Oncogene. 1993;8:307–318. [PubMed] [Google Scholar]
  • 138.Diller L, Kassel J, Nelson CE, et al. p53 functions as a cell cycle control protein in osteosarcomas. Mol Cell Biol. 1990;10:5772–5781. doi: 10.1128/mcb.10.11.5772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Henriksen R, Strang P, Wilander E, Backstrom T, Tribukait B, Oberg K. p53 expression in epithelial ovarian neoplasms: relationship to clinical and pathological parameters, Ki-67 expression and flow cytometry. Gynecol Oncol. 1994;53:301–306. doi: 10.1006/gyno.1994.1138. [DOI] [PubMed] [Google Scholar]
  • 140.Hartmann LC, Podratz KC, Keeney GL, et al. Prognostic significance of p53 immunostaining in epithelial ovarian cancer. J Clin Oncol. 1994;12:64–69. doi: 10.1200/JCO.1994.12.1.64. [DOI] [PubMed] [Google Scholar]
  • 141.Levesque MA, Katsaros D, Yu H, et al. Mutant p53 protein overexpression is associated with poor outcome in patients with well or moderately differentiated ovarian carcinoma. Cancer. 1995;75:1327–1338. doi: 10.1002/1097-0142(19950315)75:6<1327::aid-cncr2820750615>3.0.co;2-p. [DOI] [PubMed] [Google Scholar]
  • 142.Milner BJ, Allan LA, Eccles DM, et al. p53 mutation is a common genetic event in ovarian carcinoma. Cancer Res. 1993;53:2128–2132. [PubMed] [Google Scholar]
  • 143.Anttila MA, Ji H, Juhola MT, Saarikoski SV, Syrjanen KJ. The prognostic significance of p53 expression quantitated by computerized image analysis in epithelial ovarian cancer. Int J Gynecol Pathol. 1999;18:42–51. doi: 10.1097/00004347-199901000-00006. [DOI] [PubMed] [Google Scholar]
  • 144.Tammela J, Odunsi K. Gene expression and prognostic significance in ovarian cancer. Minerva Ginecol. 2004;56:495–502. [PubMed] [Google Scholar]
  • 145.Tomsova M, Melichar B. Contribution of immunohistochemistry in prognostic assessment of epithelial ovarian carcinoma --review of the literature I. Acta Medica (Hradec Kralove) 2006;49:161–165. [PubMed] [Google Scholar]
  • 146.Dobrzycka B, Terlikowski SJ, Kinalski M, Kowalczuk O, Niklinska W, Chyczewski L. Circulating free DNA and p53 antibodies in plasma of patients with ovarian epithelial cancers. Ann Oncol. doi: 10.1093/annonc/mdq584. [DOI] [PubMed] [Google Scholar]
  • 147.Goodell V, Salazar LG, Urban N, et al. Antibody immunity to the p53 oncogenic protein is a prognostic indicator in ovarian cancer. J Clin Oncol. 2006;24:762–768. doi: 10.1200/JCO.2005.03.2813. [DOI] [PubMed] [Google Scholar]
  • 148.Kihana T, Tsuda H, Teshima S, Okada S, Matsuura S, Hirohashi S. High incidence of p53 gene mutation in human ovarian cancer and its association with nuclear accumulation of p53 protein and tumor DNA aneuploidy. Jpn J Cancer Res. 1992;83:978–984. doi: 10.1111/j.1349-7006.1992.tb02010.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Zheng J, Benedict WF, Xu HJ, et al. Genetic disparity between morphologically benign cysts contiguous to ovarian carcinomas and solitary cystadenomas. J Natl Cancer Inst. 1995;87:1146–1153. doi: 10.1093/jnci/87.15.1146. [DOI] [PubMed] [Google Scholar]
  • 150.Kappes S, Milde-Langosch K, Kressin P, et al. p53 mutations in ovarian tumors, detected by temperature-gradient gel electrophoresis, direct sequencing and immunohistochemistry. Int J Cancer. 1995;64:52–59. doi: 10.1002/ijc.2910640111. [DOI] [PubMed] [Google Scholar]
  • 151.Laframboise S, Chapman W, McLaughlin J, Andrulis IL. p53 mutations in epithelial ovarian cancers: possible role in predicting chemoresistance. Cancer J. 2000;6:302–308. [PubMed] [Google Scholar]
  • 152.Rose SL, Robertson AD, Goodheart MJ, Smith BJ, DeYoung BR, Buller RE. The impact of p53 protein core domain structural alteration on ovarian cancer survival. Clin Cancer Res. 2003;9:4139–4144. [PubMed] [Google Scholar]
  • 153.Fallows S, Price J, Atkinson RJ, Johnston PG, Hickey I, Russell SE. P53 mutation does not affect prognosis in ovarian epithelial malignancies. J Pathol. 2001;194:68–75. doi: 10.1002/path.857. [DOI] [PubMed] [Google Scholar]
  • 154.Smith-Sorensen B, Kaern J, Holm R, Dorum A, Trope C, Borresen-Dale AL. Therapy effect of either paclitaxel or cyclophosphamide combination treatment in patients with epithelial ovarian cancer and relation to TP53 gene status. Br J Cancer. 1998;78:375–381. doi: 10.1038/bjc.1998.502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Niwa K, Itoh M, Murase T, et al. Alteration of p53 gene in ovarian carcinoma: clinicopathological correlation and prognostic significance. Br J Cancer. 1994;70:1191–1197. doi: 10.1038/bjc.1994.472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Wang Y, Kringen P, Kristensen GB, et al. Effect of the codon 72 polymorphism (c.215G>C, p.Arg72Pro) in combination with somatic sequence variants in the TP53 gene on survival in patients with advanced ovarian carcinoma. Hum Mutat. 2004;24:21–34. doi: 10.1002/humu.20055. [DOI] [PubMed] [Google Scholar]
  • 157.Ng SS, Cheung YT, An XM, et al. Cell cycle-related kinase: a novel candidate oncogene in human glioblastoma. J Natl Cancer Inst. 2007;99:936–948. doi: 10.1093/jnci/djm011. [DOI] [PubMed] [Google Scholar]
  • 158.Boer JM, Huber WK, Sultmann H, et al. Identification and classification of differentially expressed genes in renal cell carcinoma by expression profiling on a global human 31,500-element cDNA array. Genome Res. 2001;11:1861–1870. doi: 10.1101/gr.184501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Sherr CJ. Mammalian G1 cyclins. Cell. 1993;73:1059–1065. doi: 10.1016/0092-8674(93)90636-5. [DOI] [PubMed] [Google Scholar]
  • 160.Gillett C, Fantl V, Smith R, et al. Amplification and overexpression of cyclin D1 in breast cancer detected by immunohistochemical staining. Cancer Res. 1994;54:1812–1817. [PubMed] [Google Scholar]
  • 161.Callender T, el-Naggar AK, Lee MS, Frankenthaler R, Luna MA, Batsakis JG. PRAD-1 (CCND1)/cyclin D1 oncogene amplification in primary head and neck squamous cell carcinoma. Cancer. 1994;74:152–158. doi: 10.1002/1097-0142(19940701)74:1<152::aid-cncr2820740124>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 162.Nishida N, Fukuda Y, Komeda T, et al. Amplification and overexpression of the cyclin D1 gene in aggressive human hepatocellular carcinoma. Cancer Res. 1994;54:3107–3110. [PubMed] [Google Scholar]
  • 163.Hall M, Peters G. Genetic alterations of cyclins, cyclin-dependent kinases, and Cdk inhibitors in human cancer. Adv Cancer Res. 1996;68:67–108. doi: 10.1016/s0065-230x(08)60352-8. [DOI] [PubMed] [Google Scholar]
  • 164.Worsley SD, Ponder BA, Davies BR. Overexpression of cyclin D1 in epithelial ovarian cancers. Gynecol Oncol. 1997;64:189–195. doi: 10.1006/gyno.1996.4569. [DOI] [PubMed] [Google Scholar]
  • 165.Masciullo V, Scambia G, Marone M, et al. Altered expression of cyclin D1 and CDK4 genes in ovarian carcinomas. Int J Cancer. 1997;74:390–395. doi: 10.1002/(sici)1097-0215(19970822)74:4<390::aid-ijc5>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 166.Barbieri F, Cagnoli M, Ragni N, Pedulla F, Foglia G, Alama A. Expression of cyclin D1 correlates with malignancy in human ovarian tumours. Br J Cancer. 1997;75:1263–1268. doi: 10.1038/bjc.1997.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Bali A, O'Brien PM, Edwards LS, Sutherland RL, Hacker NF, Henshall SM. Cyclin D1, p53, and p21Waf1/Cip1 expression is predictive of poor clinical outcome in serous epithelial ovarian cancer. Clin Cancer Res. 2004;10:5168–5177. doi: 10.1158/1078-0432.CCR-03-0751. [DOI] [PubMed] [Google Scholar]
  • 168.Barbieri F, Cagnoli M, Ragni N, et al. Increased cyclin D1 expression is associated with features of malignancy and disease recurrence in ovarian tumors. Clin Cancer Res. 1999;5:1837–1842. [PubMed] [Google Scholar]
  • 169.Sui L, Tokuda M, Ohno M, Hatase O, Hando T. The concurrent expression of p27(kip1) and cyclin D1 in epithelial ovarian tumors. Gynecol Oncol. 1999;73:202–209. doi: 10.1006/gyno.1999.5373. [DOI] [PubMed] [Google Scholar]
  • 170.Chen CH, Shen J, Lee WJ, Chow SN. Overexpression of cyclin D1 and c-Myc gene products in human primary epithelial ovarian cancer. Int J Gynecol Cancer. 2005;15:878–883. doi: 10.1111/j.1525-1438.2005.00150.x. [DOI] [PubMed] [Google Scholar]
  • 171.Thangaraju M, Kaufmann SH, Couch FJ. BRCA1 facilitates stress-induced apoptosis in breast and ovarian cancer cell lines. J Biol Chem. 2000;275:33487–33496. doi: 10.1074/jbc.M005824200. [DOI] [PubMed] [Google Scholar]
  • 172.Lengauer C, Kinzler KW, Vogelstein B. Genetic instability in colorectal cancers. Nature. 1997;386:623–627. doi: 10.1038/386623a0. [DOI] [PubMed] [Google Scholar]
  • 173.Bani-Hani KE, Almasri NM, Khader YS, Sheyab FM, Karam HN. Combined evaluation of expressions of cyclin E and p53 proteins as prognostic factors for patients with gastric cancer. Clin Cancer Res. 2005;11:1447–1453. doi: 10.1158/1078-0432.CCR-04-1730. [DOI] [PubMed] [Google Scholar]
  • 174.Fukuse T, Hirata T, Naiki H, Hitomi S, Wada H. Prognostic significance of cyclin E overexpression in resected non-small cell lung cancer. Cancer Res. 2000;60:242–244. [PubMed] [Google Scholar]
  • 175.Ferreri AJ, Ponzoni M, Pruneri G, et al. Immunoreactivity for p27(KIP1) and cyclin E is an independent predictor of survival in primary gastric non-Hodgkin's lymphoma. Int J Cancer. 2001;94:599–604. doi: 10.1002/ijc.1509. [DOI] [PubMed] [Google Scholar]
  • 176.Courjal F, Louason G, Speiser P, Katsaros D, Zeillinger R, Theillet C. Cyclin gene amplification and overexpression in breast and ovarian cancers: evidence for the selection of cyclin D1 in breast and cyclin E in ovarian tumors. Int J Cancer. 1996;69:247–253. doi: 10.1002/(SICI)1097-0215(19960822)69:4<247::AID-IJC1>3.0.CO;2-X. [DOI] [PubMed] [Google Scholar]
  • 177.Farley J, Smith LM, Darcy KM, et al. Cyclin E expression is a significant predictor of survival in advanced, suboptimally debulked ovarian epithelial cancers: a Gynecologic Oncology Group study. Cancer Res. 2003;63:1235–1241. [PubMed] [Google Scholar]
  • 178.Yamamoto S, Tsuda H, Miyai K, Takano M, Tamai S, Matsubara O. Cumulative alterations of p27-related cell-cycle regulators in the development of endometriosis-associated ovarian clear cell adenocarcinoma. Histopathology. 56:740–749. doi: 10.1111/j.1365-2559.2010.03551.x. [DOI] [PubMed] [Google Scholar]
  • 179.Marone M, Scambia G, Giannitelli C, et al. Analysis of cyclin E and CDK2 in ovarian cancer: gene amplification and RNA overexpression. Int J Cancer. 1998;75:34–39. doi: 10.1002/(sici)1097-0215(19980105)75:1<34::aid-ijc6>3.0.co;2-2. [DOI] [PubMed] [Google Scholar]
  • 180.Sui L, Dong Y, Ohno M, et al. Implication of malignancy and prognosis of p27(kip1), Cyclin E, and Cdk2 expression in epithelial ovarian tumors. Gynecol Oncol. 2001;83:56–63. doi: 10.1006/gyno.2001.6308. [DOI] [PubMed] [Google Scholar]
  • 181.Todd MC, Spruill SC, Meerbrey KL. Small interference RNA-mediated suppression of overexpressed cyclin E protein restores G1/S regulation in NIH-OVCAR-3 ovarian cancer cells. Int J Oncol. 2009;35:375–380. [PubMed] [Google Scholar]
  • 182.Shimizu M, Nikaido T, Toki T, Shiozawa T, Fujii S. Clear cell carcinoma has an expression pattern of cell cycle regulatory molecules that is unique among ovarian adenocarcinomas. Cancer. 1999;85:669–677. doi: 10.1002/(sici)1097-0142(19990201)85:3<669::aid-cncr17>3.0.co;2-f. [DOI] [PubMed] [Google Scholar]
  • 183.Ohtsubo M, Roberts JM. Cyclin-dependent regulation of G1 in mammalian fibroblasts. Science. 1993;259:1908–1912. doi: 10.1126/science.8384376. [DOI] [PubMed] [Google Scholar]
  • 184.Bedrosian I, Lu KH, Verschraegen C, Keyomarsi K. Cyclin E deregulation alters the biologic properties of ovarian cancer cells. Oncogene. 2004;23:2648–2657. doi: 10.1038/sj.onc.1207408. [DOI] [PubMed] [Google Scholar]
  • 185.Akli S, Zheng PJ, Multani AS, et al. Tumor-specific low molecular weight forms of cyclin E induce genomic instability and resistance to p21, p27, and antiestrogens in breast cancer. Cancer Res. 2004;64:3198–3208. doi: 10.1158/0008-5472.can-03-3672. [DOI] [PubMed] [Google Scholar]
  • 186.Spruck CH, Won KA, Reed SI. Deregulated cyclin E induces chromosome instability. Nature. 1999;401:297–300. doi: 10.1038/45836. [DOI] [PubMed] [Google Scholar]
  • 187.Rosen DG, Yang G, Deavers MT, et al. Cyclin E expression is correlated with tumor progression and predicts a poor prognosis in patients with ovarian carcinoma. Cancer. 2006;106:1925–1932. doi: 10.1002/cncr.21767. [DOI] [PubMed] [Google Scholar]
  • 188.Blegen H, Einhorn N, Sjovall K, et al. Prognostic significance of cell cycle proteins and genomic instability in borderline, early and advanced stage ovarian carcinomas. Int J Gynecol Cancer. 2000;10:477–487. doi: 10.1046/j.1525-1438.2000.00077.x. [DOI] [PubMed] [Google Scholar]
  • 189.Milde-Langosch K, Hagen M, Bamberger AM, Loning T. Expression and prognostic value of the cell-cycle regulatory proteins, Rb, p16MTS1, p21WAF1, p27KIP1, cyclin E, and cyclin D2, in ovarian cancer. Int J Gynecol Pathol. 2003;22:168–174. doi: 10.1097/00004347-200304000-00009. [DOI] [PubMed] [Google Scholar]
  • 190.Scully R, Livingston DM. In search of the tumour-suppressor functions of BRCA1 and BRCA2. Nature. 2000;408:429–432. doi: 10.1038/35044000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Leegte B, van der Hout AH, Deffenbaugh AM, et al. Phenotypic expression of double heterozygosity for BRCA1 and BRCA2 germline mutations. J Med Genet. 2005;42:e20. doi: 10.1136/jmg.2004.027243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Warner E, Foulkes W, Goodwin P, et al. Prevalence and penetrance of BRCA1 and BRCA2 gene mutations in unselected Ashkenazi Jewish women with breast cancer. J Natl Cancer Inst. 1999;91:1241–1247. doi: 10.1093/jnci/91.14.1241. [DOI] [PubMed] [Google Scholar]
  • 193.Piura B, Rabinovich A, Yanai-Inbar I. Three primary malignancies related to BRCA mutation successively occurring in a BRCA1 185delAG mutation carrier. Eur J Obstet Gynecol Reprod Biol. 2001;97:241–244. doi: 10.1016/s0301-2115(00)00521-2. [DOI] [PubMed] [Google Scholar]
  • 194.Ford D, Easton DF, Bishop DT, Narod SA, Goldgar DE. Risks of cancer in BRCA1-mutation carriers. Breast Cancer Linkage Consortium. Lancet. 1994;343:692–695. doi: 10.1016/s0140-6736(94)91578-4. [DOI] [PubMed] [Google Scholar]
  • 195.Ford D, Easton DF, Stratton M, et al. Genetic heterogeneity and penetrance analysis of the BRCA1 and BRCA2 genes in breast cancer families. The Breast Cancer Linkage Consortium. Am J Hum Genet. 1998;62:676–689. doi: 10.1086/301749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Matzuk MM. Gynecologic diseases get their genes. Nat Med. 2005;11:24–26. doi: 10.1038/nm0105-24. [DOI] [PubMed] [Google Scholar]
  • 197.Rubin SC, Benjamin I, Behbakht K, et al. Clinical and pathological features of ovarian cancer in women with germ-line mutations of BRCA1. N Engl J Med. 1996;335:1413–1416. doi: 10.1056/NEJM199611073351901. [DOI] [PubMed] [Google Scholar]
  • 198.Aida H, Takakuwa K, Nagata H, et al. Clinical features of ovarian cancer in Japanese women with germ-line mutations of BRCA1. Clin Cancer Res. 1998;4:235–240. [PubMed] [Google Scholar]
  • 199.Boyd J, Sonoda Y, Federici MG, et al. Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. JAMA. 2000;283:2260–2265. doi: 10.1001/jama.283.17.2260. [DOI] [PubMed] [Google Scholar]
  • 200.Ben David Y, Chetrit A, Hirsh-Yechezkel G, et al. Effect of BRCA mutations on the length of survival in epithelial ovarian tumors. J Clin Oncol. 2002;20:463–466. doi: 10.1200/JCO.2002.20.2.463. [DOI] [PubMed] [Google Scholar]
  • 201.Majdak EJ, Debniak J, Milczek T, et al. Prognostic impact of BRCA1 pathogenic and BRCA1/BRCA2 unclassified variant mutations in patients with ovarian carcinoma. Cancer. 2005;104:1004–1012. doi: 10.1002/cncr.21276. [DOI] [PubMed] [Google Scholar]
  • 202.Chetrit A, Hirsh-Yechezkel G, Ben-David Y, Lubin F, Friedman E, Sadetzki S. Effect of BRCA1/2 mutations on long-term survival of patients with invasive ovarian cancer: the national Israeli study of ovarian cancer. J Clin Oncol. 2008;26:20–25. doi: 10.1200/JCO.2007.11.6905. [DOI] [PubMed] [Google Scholar]
  • 203.Tan DS, Rothermundt C, Thomas K, et al. "BRCAness" syndrome in ovarian cancer: a case-control study describing the clinical features and outcome of patients with epithelial ovarian cancer associated with BRCA1 and BRCA2 mutations. J Clin Oncol. 2008;26:5530–5536. doi: 10.1200/JCO.2008.16.1703. [DOI] [PubMed] [Google Scholar]
  • 204.Frumovitz M, Schmeler KM, Malpica A, Sood AK, Gershenson DM. Unmasking the complexities of mucinous ovarian carcinoma. Gynecol Oncol. 117:491–496. doi: 10.1016/j.ygyno.2010.02.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Ramus SJ, Fishman A, Pharoah PD, Yarkoni S, Altaras M, Ponder BA. Ovarian cancer survival in Ashkenazi Jewish patients with BRCA1 and BRCA2 mutations. Eur J Surg Oncol. 2001;27:278–281. doi: 10.1053/ejso.2000.1097. [DOI] [PubMed] [Google Scholar]
  • 206.Cass I, Baldwin RL, Varkey T, Moslehi R, Narod SA, Karlan BY. Improved survival in women with BRCA-associated ovarian carcinoma. Cancer. 2003;97:2187–2195. doi: 10.1002/cncr.11310. [DOI] [PubMed] [Google Scholar]
  • 207.Dabholkar M, Vionnet J, Bostick-Bruton F, Yu JJ, Reed E. Messenger RNA levels of XPAC and ERCC1 in ovarian cancer tissue correlate with response to platinum-based chemotherapy. J Clin Invest. 1994;94:703–708. doi: 10.1172/JCI117388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Walsh CS, Ogawa S, Karahashi H, et al. ERCC5 is a novel biomarker of ovarian cancer prognosis. J Clin Oncol. 2008;26:2952–2958. doi: 10.1200/JCO.2007.13.5806. [DOI] [PubMed] [Google Scholar]
  • 209.Ceppi P, Volante M, Novello S, et al. ERCC1 and RRM1 gene expressions but not EGFR are predictive of shorter survival in advanced non-small-cell lung cancer treated with cisplatin and gemcitabine. Ann Oncol. 2006;17:1818–1825. doi: 10.1093/annonc/mdl300. [DOI] [PubMed] [Google Scholar]
  • 210.Reed E, Yu JJ, Davies A, Gannon J, Armentrout SL. Clear cell tumors have higher mRNA levels of ERCC1 and XPB than other histological types of epithelial ovarian cancer. Clin Cancer Res. 2003;9:5299–5305. [PubMed] [Google Scholar]
  • 211.Selvakumaran M, Pisarcik DA, Bao R, Yeung AT, Hamilton TC. Enhanced cisplatin cytotoxicity by disturbing the nucleotide excision repair pathway in ovarian cancer cell lines. Cancer Res. 2003;63:1311–1316. [PubMed] [Google Scholar]
  • 212.Weberpals J, Garbuio K, O'Brien A, et al. The DNA repair proteins BRCA1 and ERCC1 as predictive markers in sporadic ovarian cancer. Int J Cancer. 2009;124:806–815. doi: 10.1002/ijc.23987. [DOI] [PubMed] [Google Scholar]
  • 213.Lin K, Ye D, Xie X. Protein expression levels of excision repair cross-complementation group 1 and xeroderma pigmentosum D correlate with response to platinum-based chemotherapy in the patients with advanced epithelial ovarian cancer. Int J Gynecol Cancer. 2008;18:1007–1012. doi: 10.1111/j.1525-1438.2007.01155.x. [DOI] [PubMed] [Google Scholar]
  • 214.Shahzad MM, Lopez-Berestein G, Sood AK. Novel strategies for reversing platinum resistance. Drug Resist Updat. 2009;12:148–152. doi: 10.1016/j.drup.2009.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Aletti GD, Gostout BS, Podratz KC, Cliby WA. Ovarian cancer surgical resectability: relative impact of disease, patient status, and surgeon. Gynecol Oncol. 2006;100:33–37. doi: 10.1016/j.ygyno.2005.07.123. [DOI] [PubMed] [Google Scholar]
  • 216.Fader AN, Rose PG. Role of surgery in ovarian carcinoma. J Clin Oncol. 2007;25:2873–2883. doi: 10.1200/JCO.2007.11.0932. [DOI] [PubMed] [Google Scholar]
  • 217.Swann JB, Smyth MJ. Immune surveillance of tumors. J Clin Invest. 2007;117:1137–1146. doi: 10.1172/JCI31405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Zitvogel L, Tesniere A, Kroemer G. Cancer despite immunosurveillance: immunoselection and immunosubversion. Nat Rev Immunol. 2006;6:715–727. doi: 10.1038/nri1936. [DOI] [PubMed] [Google Scholar]
  • 219.Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213. doi: 10.1056/NEJMoa020177. [DOI] [PubMed] [Google Scholar]
  • 220.Han LY, Fletcher MS, Urbauer DL, et al. HLA class I antigen processing machinery component expression and intratumoral T-Cell infiltrate as independent prognostic markers in ovarian carcinoma. Clin Cancer Res. 2008;14:3372–3379. doi: 10.1158/1078-0432.CCR-07-4433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Zang X, Sullivan PS, Soslow RA, et al. Tumor associated endothelial expression of B7-H3 predicts survival in ovarian carcinomas. Mod Pathol. 23:1104–1112. doi: 10.1038/modpathol.2010.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Lee RC, Feinbaum RL, Ambros V. The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell. 1993;75:843–854. doi: 10.1016/0092-8674(93)90529-y. [DOI] [PubMed] [Google Scholar]
  • 223.Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 2004;116:281–297. doi: 10.1016/s0092-8674(04)00045-5. [DOI] [PubMed] [Google Scholar]
  • 224.He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet. 2004;5:522–531. doi: 10.1038/nrg1379. [DOI] [PubMed] [Google Scholar]
  • 225.Ambros V. microRNAs: tiny regulators with great potential. Cell. 2001;107:823–826. doi: 10.1016/s0092-8674(01)00616-x. [DOI] [PubMed] [Google Scholar]
  • 226.van Jaarsveld MT, Helleman J, Berns EM, Wiemer EA. MicroRNAs in ovarian cancer biology and therapy resistance. Int J Biochem Cell Biol. 42:1282–1290. doi: 10.1016/j.biocel.2010.01.014. [DOI] [PubMed] [Google Scholar]
  • 227.Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T. Impaired microRNA processing enhances cellular transformation and tumorigenesis. Nat Genet. 2007;39:673–677. doi: 10.1038/ng2003. [DOI] [PubMed] [Google Scholar]
  • 228.Calin GA, Croce CM. MicroRNA-cancer connection: the beginning of a new tale. Cancer Res. 2006;66:7390–7394. doi: 10.1158/0008-5472.CAN-06-0800. [DOI] [PubMed] [Google Scholar]
  • 229.Hu Y, Correa AM, Hoque A, et al. Prognostic significance of differentially expressed miRNAs in esophageal cancer. Int J Cancer. 128:132–143. doi: 10.1002/ijc.25330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Zhang L, Huang J, Yang N, et al. microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci U S A. 2006;103:9136–9141. doi: 10.1073/pnas.0508889103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Yang H, Kong W, He L, et al. MicroRNA expression profiling in human ovarian cancer: miR-214 induces cell survival and cisplatin resistance by targeting PTEN. Cancer Res. 2008;68:425–433. doi: 10.1158/0008-5472.CAN-07-2488. [DOI] [PubMed] [Google Scholar]
  • 232.Nam EJ, Yoon H, Kim SW, et al. MicroRNA expression profiles in serous ovarian carcinoma. Clin Cancer Res. 2008;14:2690–2695. doi: 10.1158/1078-0432.CCR-07-1731. [DOI] [PubMed] [Google Scholar]
  • 233.Kim VN. MicroRNA biogenesis: coordinated cropping and dicing. Nat Rev Mol Cell Biol. 2005;6:376–385. doi: 10.1038/nrm1644. [DOI] [PubMed] [Google Scholar]
  • 234.Croce CM. Causes and consequences of microRNA dysregulation in cancer. Nat Rev Genet. 2009;10:704–714. doi: 10.1038/nrg2634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Medina PP, Slack FJ. microRNAs and cancer: an overview. Cell Cycle. 2008;7:2485–2492. doi: 10.4161/cc.7.16.6453. [DOI] [PubMed] [Google Scholar]
  • 236.Negrini M, Nicoloso MS, Calin GA. MicroRNAs and cancer--new paradigms in molecular oncology. Curr Opin Cell Biol. 2009;21:470–479. doi: 10.1016/j.ceb.2009.03.002. [DOI] [PubMed] [Google Scholar]
  • 237.Shah PP, Hutchinson LE, Kakar SS. Emerging role of microRNAs in diagnosis and treatment of various diseases including ovarian cancer. J Ovarian Res. 2009;2:11. doi: 10.1186/1757-2215-2-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Merritt WM, Lin YG, Han LY, et al. Dicer, Drosha, and outcomes in patients with ovarian cancer. N Engl J Med. 2008;359:2641–2650. doi: 10.1056/NEJMoa0803785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Chiosea S, Jelezcova E, Chandran U, et al. Up-regulation of dicer, a component of the MicroRNA machinery, in prostate adenocarcinoma. Am J Pathol. 2006;169:1812–1820. doi: 10.2353/ajpath.2006.060480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Faggad A, Budczies J, Tchernitsa O, et al. Prognostic significance of Dicer expression in ovarian cancer-link to global microRNA changes and oestrogen receptor expression. J Pathol. 220:382–391. doi: 10.1002/path.2658. [DOI] [PubMed] [Google Scholar]
  • 241.Wu JF, Shen W, Liu NZ, et al. Down-regulation of Dicer in hepatocellular carcinoma. Med Oncol. doi: 10.1007/s12032-010-9520-5. [DOI] [PubMed] [Google Scholar]
  • 242.Martin MG, Payton JE, Link DC. Dicer and outcomes in patients with acute myeloid leukemia (AML) Leuk Res. 2009;33:e127. doi: 10.1016/j.leukres.2009.02.003. [DOI] [PubMed] [Google Scholar]
  • 243.Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K. Modulation of microRNA processing by p53. Nature. 2009;460:529–533. doi: 10.1038/nature08199. [DOI] [PubMed] [Google Scholar]
  • 244.Stroun M, Maurice P, Vasioukhin V, et al. The origin and mechanism of circulating DNA. Ann N Y Acad Sci. 2000;906:161–168. doi: 10.1111/j.1749-6632.2000.tb06608.x. [DOI] [PubMed] [Google Scholar]
  • 245.Anker P, Mulcahy H, Chen XQ, Stroun M. Detection of circulating tumour DNA in the blood (plasma/serum) of cancer patients. Cancer Metastasis Rev. 1999;18:65–73. doi: 10.1023/a:1006260319913. [DOI] [PubMed] [Google Scholar]
  • 246.Mandel P, Metais P. [Not Available.] C R Seances Soc Biol Fil. 1948;142:241–243. [PubMed] [Google Scholar]
  • 247.Fournie GJ, Courtin JP, Laval F, et al. Plasma DNA as a marker of cancerous cell death. Investigations in patients suffering from lung cancer and in nude mice bearing human tumours. Cancer Lett. 1995;91:221–227. doi: 10.1016/0304-3835(95)03742-f. [DOI] [PubMed] [Google Scholar]
  • 248.Holdenrieder S, Stieber P, Forg T, et al. Apoptosis in serum of patients with solid tumours. Anticancer Res. 1999;19:2721–2724. [PubMed] [Google Scholar]
  • 249.Stroun M, Anker P, Lyautey J, Lederrey C, Maurice PA. Isolation and characterization of DNA from the plasma of cancer patients. Eur J Cancer Clin Oncol. 1987;23:707–712. doi: 10.1016/0277-5379(87)90266-5. [DOI] [PubMed] [Google Scholar]
  • 250.Chen X, Bonnefoi H, Diebold-Berger S, et al. Detecting tumor-related alterations in plasma or serum DNA of patients diagnosed with breast cancer. Clin Cancer Res. 1999;5:2297–2303. [PubMed] [Google Scholar]
  • 251.Nawroz H, Koch W, Anker P, Stroun M, Sidransky D. Microsatellite alterations in serum DNA of head and neck cancer patients. Nat Med. 1996;2:1035–1037. doi: 10.1038/nm0996-1035. [DOI] [PubMed] [Google Scholar]
  • 252.Fiegl H, Millinger S, Mueller-Holzner E, et al. Circulating tumor-specific DNA: a marker for monitoring efficacy of adjuvant therapy in cancer patients. Cancer Res. 2005;65:1141–1145. doi: 10.1158/0008-5472.CAN-04-2438. [DOI] [PubMed] [Google Scholar]
  • 253.Kamat AA, Bischoff FZ, Dang D, et al. Circulating cell-free DNA: a novel biomarker for response to therapy in ovarian carcinoma. Cancer Biol Ther. 2006;5:1369–1374. doi: 10.4161/cbt.5.10.3240. [DOI] [PubMed] [Google Scholar]
  • 254.Wei WI, Yuen AP, Ng RW, Ho WK, Kwong DL, Sham JS. Quantitative analysis of plasma cell-free Epstein-Barr virus DNA in nasopharyngeal carcinoma after salvage nasopharyngectomy: a prospective study. Head Neck. 2004;26:878–883. doi: 10.1002/hed.20066. [DOI] [PubMed] [Google Scholar]
  • 255.Gautschi O, Bigosch C, Huegli B, et al. Circulating deoxyribonucleic Acid as prognostic marker in non-small-cell lung cancer patients undergoing chemotherapy. J Clin Oncol. 2004;22:4157–4164. doi: 10.1200/JCO.2004.11.123. [DOI] [PubMed] [Google Scholar]
  • 256.Leung SF, Chan AT, Zee B, et al. Pretherapy quantitative measurement of circulating Epstein-Barr virus DNA is predictive of posttherapy distant failure in patients with early-stage nasopharyngeal carcinoma of undifferentiated type. Cancer. 2003;98:288–291. doi: 10.1002/cncr.11496. [DOI] [PubMed] [Google Scholar]
  • 257.Zachariah RR, Schmid S, Buerki N, Radpour R, Holzgreve W, Zhong X. Levels of circulating cell-free nuclear and mitochondrial DNA in benign and malignant ovarian tumors. Obstet Gynecol. 2008;112:843–850. doi: 10.1097/AOG.0b013e3181867bc0. [DOI] [PubMed] [Google Scholar]
  • 258.Moore RG, Brown AK, Miller MC, et al. The use of multiple novel tumor biomarkers for the detection of ovarian carcinoma in patients with a pelvic mass. Gynecol Oncol. 2008;108:402–408. doi: 10.1016/j.ygyno.2007.10.017. [DOI] [PubMed] [Google Scholar]
  • 259.Lu KH, Patterson AP, Wang L, et al. Selection of potential markers for epithelial ovarian cancer with gene expression arrays and recursive descent partition analysis. Clin Cancer Res. 2004;10:3291–3300. doi: 10.1158/1078-0432.CCR-03-0409. [DOI] [PubMed] [Google Scholar]
  • 260.McIntosh MW, Drescher C, Karlan B, et al. Combining CA 125 and SMR serum markers for diagnosis and early detection of ovarian carcinoma. Gynecol Oncol. 2004;95:9–15. doi: 10.1016/j.ygyno.2004.07.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Jacobs IJ, Rivera H, Oram DH, Bast RC., Jr Differential diagnosis of ovarian cancer with tumour markers CA 125, CA 15-3 and TAG 72.3. Br J Obstet Gynaecol. 1993;100:1120–1124. doi: 10.1111/j.1471-0528.1993.tb15177.x. [DOI] [PubMed] [Google Scholar]
  • 262.Li C, Wong WH. Model-based analysis of oligonucleotide arrays: expression index computation and outlier detection. Proc Natl Acad Sci U S A. 2001;98:31–36. doi: 10.1073/pnas.011404098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 263.Chau CH, Rixe O, McLeod H, Figg WD. Validation of analytic methods for biomarkers used in drug development. Clin Cancer Res. 2008;14:5967–5976. doi: 10.1158/1078-0432.CCR-07-4535. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 264.Hogdall E. Cancer antigen 125 and prognosis. Curr Opin Obstet Gynecol. 2008;20:4–8. doi: 10.1097/GCO.0b013e3282f2b124. [DOI] [PubMed] [Google Scholar]
  • 265.Altman DG, Lyman GH. Methodological challenges in the evaluation of prognostic factors in breast cancer. Breast Cancer Res Treat. 1998;52:289–303. doi: 10.1023/a:1006193704132. [DOI] [PubMed] [Google Scholar]
  • 266.Hayes DF, Bast RC, Desch CE, et al. Tumor marker utility grading system: a framework to evaluate clinical utility of tumor markers. J Natl Cancer Inst. 1996;88:1456–1466. doi: 10.1093/jnci/88.20.1456. [DOI] [PubMed] [Google Scholar]
  • 267.McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. REporting recommendations for tumor MARKer prognostic studies (REMARK) Nat Clin Pract Oncol. 2005;2:416–422. [PubMed] [Google Scholar]
  • 268.Cheng WF, Huang CY, Chang MC, et al. High mesothelin correlates with chemoresistance and poor survival in epithelial ovarian carcinoma. Br J Cancer. 2009;100:1144–1153. doi: 10.1038/sj.bjc.6604964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Katz EM, Kattan MW. How to judge a tumor marker. Nat Clin Pract Oncol. 2005;2:482–483. doi: 10.1038/ncponc0318. [DOI] [PubMed] [Google Scholar]
  • 270.Liotta LA, Petricoin EF. Mass spectrometry-based protein biomarker discovery: solving the remaining challenges to reach the promise of clinical benefit. Clin Chem. 56:1641–1642. doi: 10.1373/clinchem.2010.146142. [DOI] [PubMed] [Google Scholar]
  • 271.Khleif SN, Doroshow JH, Hait WN. AACR-FDA-NCI Cancer Biomarkers Collaborative consensus report: advancing the use of biomarkers in cancer drug development. Clin Cancer Res. 16:3299–3318. doi: 10.1158/1078-0432.CCR-10-0880. [DOI] [PubMed] [Google Scholar]
  • 272.Abdel-Azeez HA, Labib HA, Sharaf SM, Refai AN. HE4 and mesothelin: novel biomarkers of ovarian carcinoma in patients with pelvic masses. Asian Pac J Cancer Prev. 11:111–116. [PubMed] [Google Scholar]
  • 273.Huang CY, Cheng WF, Lee CN, et al. Serum mesothelin in epithelial ovarian carcinoma: a new screening marker and prognostic factor. Anticancer Res. 2006;26:4721–4728. [PubMed] [Google Scholar]
  • 274.Chambers SK, Kacinski BM, Ivins CM, Carcangiu ML. Overexpression of epithelial macrophage colony-stimulating factor (CSF-1) and CSF-1 receptor: a poor prognostic factor in epithelial ovarian cancer, contrasted with a protective effect of stromal CSF-1. Clin Cancer Res. 1997;3:999–1007. [PubMed] [Google Scholar]
  • 275.Bao LH, Sakaguchi H, Fujimoto J, Tamaya T. Osteopontin in metastatic lesions as a prognostic marker in ovarian cancers. J Biomed Sci. 2007;14:373–381. doi: 10.1007/s11373-006-9143-1. [DOI] [PubMed] [Google Scholar]
  • 276.White NM, Mathews M, Yousef GM, et al. Human kallikrein related peptidases 6 and 13 in combination withCA125 is a more sensitive test for ovarian cancer than CA125 alone. Cancer Biomark. 2009;5:279–287. doi: 10.3233/CBM-2009-0113. [DOI] [PubMed] [Google Scholar]
  • 277.Matsuzaki H, Kobayashi H, Yagyu T, et al. Plasma bikunin as a favorable prognostic factor in ovarian cancer. J Clin Oncol. 2005;23:1463–1472. doi: 10.1200/JCO.2005.03.010. [DOI] [PubMed] [Google Scholar]
  • 278.Hefler LA, Zeillinger R, Grimm C, et al. Preoperative serum vascular endothelial growth factor as a prognostic parameter in ovarian cancer. Gynecol Oncol. 2006;103:512–517. doi: 10.1016/j.ygyno.2006.03.058. [DOI] [PubMed] [Google Scholar]
  • 279.Rudlowski C, Pickart AK, Fuhljahn C, et al. Prognostic significance of vascular endothelial growth factor expression in ovarian cancer patients: a long-term follow-up. Int J Gynecol Cancer. 2006;16(Suppl 1):183–189. doi: 10.1111/j.1525-1438.2006.00307.x. [DOI] [PubMed] [Google Scholar]
  • 280.Polterauer S, Grimm C, Mustea A, et al. Vascular endothelial growth factor gene polymorphisms in ovarian cancer. Gynecol Oncol. 2007;105:385–389. doi: 10.1016/j.ygyno.2006.12.021. [DOI] [PubMed] [Google Scholar]
  • 281.Birrer MJ, Johnson ME, Hao K, et al. Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas. J Clin Oncol. 2007;25:2281–2287. doi: 10.1200/JCO.2006.09.0795. [DOI] [PubMed] [Google Scholar]
  • 282.Li H, Cai KQ, Godwin AK, Zhang R. Enhancer of zeste homolog 2 (EZH2) promotes the proliferation and invasion of epithelial ovarian cancer cells. Mol Cancer Res. doi: 10.1158/1541-7786.MCR-10-0398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Kleinberg L, Holth A, Trope CG, Reich R, Davidson B. Claudin upregulation in ovarian carcinoma effusions is associated with poor survival. Hum Pathol. 2008;39:747–757. doi: 10.1016/j.humpath.2007.10.002. [DOI] [PubMed] [Google Scholar]
  • 284.Lin CK, Chao TK, Yu CP, Yu MH, Jin JS. The expression of six biomarkers in the four most common ovarian cancers: correlation with clinicopathological parameters. APMIS. 2009;117:162–175. doi: 10.1111/j.1600-0463.2008.00003.x. [DOI] [PubMed] [Google Scholar]
  • 285.Steffensen KD, Waldstrom M, Jeppesen U, Jakobsen E, Brandslund I, Jakobsen A. The prognostic importance of cyclooxygenase 2 and HER2 expression in epithelial ovarian cancer. Int J Gynecol Cancer. 2007;17:798–807. doi: 10.1111/j.1525-1438.2006.00855.x. [DOI] [PubMed] [Google Scholar]
  • 286.Rubin SC, Finstad CL, Wong GY, Almadrones L, Plante M, Lloyd KO. Prognostic significance of HER-2/neu expression in advanced epithelial ovarian cancer: a multivariate analysis. Am J Obstet Gynecol. 1993;168:162–169. doi: 10.1016/s0002-9378(12)90907-2. [DOI] [PubMed] [Google Scholar]
  • 287.Anderson KS, Wong J, Vitonis A, et al. p53 autoantibodies as potential detection and prognostic biomarkers in serous ovarian cancer. Cancer Epidemiol Biomarkers Prev. 19:859–868. doi: 10.1158/1055-9965.EPI-09-0880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 288.Hogdall EV, Hogdall CK, Blaakaer J, et al. P53 autoantibodies in sera from Danish ovarian cancer patients and their correlation with clinical data and prognosis. APMIS. 2002;110:545–553. doi: 10.1034/j.1600-0463.2002.11007805.x. [DOI] [PubMed] [Google Scholar]
  • 289.Barbieri F, Lorenzi P, Ragni N, et al. Overexpression of cyclin D1 is associated with poor survival in epithelial ovarian cancer. Oncology. 2004;66:310–315. doi: 10.1159/000078332. [DOI] [PubMed] [Google Scholar]
  • 290.Scheil-Bertram S, Tylus-Schaaf P, du Bois A, et al. Excision repair cross-complementation group 1 protein overexpression as a predictor of poor survival for high-grade serous ovarian adenocarcinoma. Gynecol Oncol. 119:325–331. doi: 10.1016/j.ygyno.2010.07.018. [DOI] [PubMed] [Google Scholar]
  • 291.Lin M, Yin N, Murphy B, et al. Immune cell-derived c3 is required for autoimmune diabetes induced by multiple low doses of streptozotocin. Diabetes. 59:2247–2252. doi: 10.2337/db10-0044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 292.Lin F, Lin K, Xie X, Zhou C. Increased ERCC1 protein expression is associated with suboptimal debulking in advanced epithelial ovarian cancer. Anticancer Res. 30:2447–2452. [PubMed] [Google Scholar]
  • 293.Konstantinopoulos PA, Spentzos D, Karlan BY, et al. Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer. J Clin Oncol. 28:3555–3561. doi: 10.1200/JCO.2009.27.5719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Cvelbar M, Ursic-Vrscaj M, Rakar S. Risk factors and prognostic factors in patients with double primary cancer: epithelial ovarian cancer and breast cancer. Eur J Gynaecol Oncol. 2005;26:59–63. [PubMed] [Google Scholar]
  • 295.Penson RT, Kronish K, Duan Z, et al. Cytokines IL-1beta, IL-2, IL-6, IL-8, MCP-1, GM-CSF and TNFalpha in patients with epithelial ovarian cancer and their relationship to treatment with paclitaxel. Int J Gynecol Cancer. 2000;10:33–41. doi: 10.1046/j.1525-1438.2000.00003.x. [DOI] [PubMed] [Google Scholar]
  • 296.Lambeck AJ, Crijns AP, Leffers N, et al. Serum cytokine profiling as a diagnostic and prognostic tool in ovarian cancer: a potential role for interleukin 7. Clin Cancer Res. 2007;13:2385–2391. doi: 10.1158/1078-0432.CCR-06-1828. [DOI] [PubMed] [Google Scholar]
  • 297.Zeimet AG, Widschwendter M, Knabbe C, et al. Ascitic interleukin-12 is an independent prognostic factor in ovarian cancer. J Clin Oncol. 1998;16:1861–1868. doi: 10.1200/JCO.1998.16.5.1861. [DOI] [PubMed] [Google Scholar]
  • 298.Cheng X, Jiang R, Li ZT, et al. The role of secondary cytoreductive surgery for recurrent mucinous epithelial ovarian cancer (mEOC) Eur J Surg Oncol. 2009;35:1105–1108. doi: 10.1016/j.ejso.2009.03.010. [DOI] [PubMed] [Google Scholar]
  • 299.Simon I, Katsaros D, Rigault de la Longrais I, et al. B7-H4 is over-expressed in early-stage ovarian cancer and is independent of CA125 expression. Gynecol Oncol. 2007;106:334–341. doi: 10.1016/j.ygyno.2007.03.035. [DOI] [PubMed] [Google Scholar]
  • 300.Marth C, Fiegl H, Zeimet AG, et al. Interferon-gamma expression is an independent prognostic factor in ovarian cancer. Am J Obstet Gynecol. 2004;191:1598–1605. doi: 10.1016/j.ajog.2004.05.007. [DOI] [PubMed] [Google Scholar]
  • 301.Duncan TJ, Rolland P, Deen S, et al. Loss of IFN gamma receptor is an independent prognostic factor in ovarian cancer. Clin Cancer Res. 2007;13:4139–4145. doi: 10.1158/1078-0432.CCR-06-2833. [DOI] [PubMed] [Google Scholar]
  • 302.Hu X, Macdonald DM, Huettner PC, et al. A miR-200 microRNA cluster as prognostic marker in advanced ovarian cancer. Gynecol Oncol. 2009;114:457–464. doi: 10.1016/j.ygyno.2009.05.022. [DOI] [PubMed] [Google Scholar]

RESOURCES