Biomarker discovery in ovarian cancer - Future Medicine

4 downloads 97 Views 391KB Size Report
Biomarker discovery in ovarian cancer – REVIEW. 29 future science group www.futuremedicine.com ratio between the test and reference samples for each of ...
R EVIEW

For reprint orders, please contact: [email protected]

Biomarker discovery in ovarian cancer Celestine S Tung1, Kwong-Kwok Wong2 & Samuel C Mok3† †Author

for correspondence of Texas, MD Anderson Cancer Center, Department of Gynecologic Oncology, 1515 Holcombe Blvd, Unit 1362, Houston, TX 77030, USA Tel.: +1 713 563 4562; Fax: +1 713 792 7586; E-mail: ctung@ mdanderson.org 2Tel.: +1 713 792 0229; Fax: +1 713 792 7586; E-mail: kkwong@ mdanderson.org 3Harvard Medical School, Department of Obstetrics, Gynecology, and Reproductive Biology, Division of Gynecologic Oncology, Brigham and Women’s Hospital, 221 Longwood Avenue, BLI-449, Boston, MA 02115, USA Tel.: +1 617 278 0196; Fax: +1 617 975 0818; E-mail: scmok@ rics.bwh.harvard.edu 1University

Keywords: biomarker, genomics, ovarian cancer, proteomics part of

Ovarian cancer is a leading cause of gynecologic cancer death among women. Tumors diagnosed early (in stage I) have a cure rate approaching 90%. However, because specific symptoms and screening tools are lacking, most ovarian cancers are very advanced when finally diagnosed. CA125 expression and pelvic ultrasonography are of limited efficacy in screening, and the search for new, complementary ovarian cancer biomarkers continues. New technology and research techniques have allowed the identification of over 100 possible tumor markers, many of which are still being evaluated for clinical relevance and several of which have entered clinical trials. Here, we review the methods of biomarker discovery, address the significance and functions of newly identified ovarian cancer tumor markers, and provide further insight into the future of ovarian cancer biomarkers.

Ovarian cancer is the deadliest gynecologic malignancy and a leading cause of cancer death among women. In the USA, an estimated 22,430 new cases will be diagnosed and 15,280 women will die of the disease in 2007 [201]. The cure rate for ovarian cancer when diagnosed in stage I approaches 90%; however, fewer than 25% of tumors are diagnosed in this early stage [1]. Over 50% of ovarian cancer patients survive for 5 years, but the cure rate for those with advanced disease is less than 30%. Therefore, early detection can have a strong effect on survival. Currently, there is no recognized model of screening, since available screening methods lack the sensitivity and specificity required for accurate diagnosis. As a result, many new biomarkers are emerging as possible candidates for use in the diagnosis and treatment response of ovarian cancer. Biomarkers can be defined as any anatomic, physiologic, biochemical or molecular parameter utilized to measure disease progression or treatment response. They can be detected through physical examination, laboratory assays or medical imaging. The NIH defines a biomarker as ‘a characteristic that is objectively measured and evaluated as an indicator of normal biologic or pathogenic processes or pharmacologic responses to a therapeutic intervention’ [2]. By 2005, an estimated 30 new biomarkers with the potential to be used alone or in conjunction with CA125 had been identified [3]. Since then, over 100 promising candidates have been identified. New studies are being performed to determine potential clinical application and functionality of these markers. In this

10.2217/17455057.4.1.27 © 2008 Future Medicine Ltd ISSN 1745-5057

review, we discuss the challenges and techniques of biomarker discovery and update what has been learned regarding these novel epithelial ovarian cancer biomarkers over the last 2 years. Screening biomarker criteria Biomarkers are clinically useful for diagnosing early-stage ovarian cancer, monitoring disease progression and treatment response, and evaluating for disease recurrence. However, to ensure widespread applicability, new diagnostic biomarkers must meet three important criteria [4,5]. First, they should be noninvasive and inexpensive so as to justify their use in screening the general population. This has made especially attractive the search for biomarkers secreted specifically by tumor tissues or immune markers detected in body fluids (e.g., serum or urine). Second, screening biomarkers should be highly sensitive for early-stage disease. Most of the current ovarian cancer biomarkers are not. Third, they must have high specificity and positive predictive value (PPV). Patients who receive falsepositive results may be subjected to unnecessary tests and costly and invasive procedures, while those who receive false-negative results may have more advance disease when it is finally diagnosed, thus affecting their prognoses. Therefore, it has been recommended that all new ovarian cancer screening biomarkers should have a minimum PPV of 10% and a minimum specificity of 99.6% [4]. Populations at higher risk, including patients with a strong family history of ovarian or breast cancer, may accept a screening method of lower specificity. Women's Health (2008) 4(1), 27–40

27

REVIEW – Tung, Wong & Mok

Tools for identifying for ovarian cancer biomarkers As there is substantial heterogeneity among the various ovarian cancer subtypes, the search for new biomarkers is extremely complicated. However, this complexity has been eased somewhat by the advent of new technology (e.g., in genomics and proteomics) that has allowed the identification of many novel markers over the past few years.

Comparative genomic hybridization analysis

Genomic technology

Genomic technology involves the analysis of whole-genome chromosomal aberrations and the measurement of the expression level of thousands of genes. Multiple technologies have been developed recently for genomic analyses. These include loss of heterozygosity (LOH) analysis, comparative genomic hybridization (CGH), spectrokaryotyping (SKY), serial analysis of gene expression (SAGE), and transcriptome profiling by oligo and cDNA microarrays. Such analyses have facilitated the rapid discovery of the molecular signatures of tumors. Many of these molecular signatures are potential biomarkers. These technologies have been applied to identify genomic changes in ovarian cancer and some of them are summarized as follows. Loss of heterozygosity analysis

LOH occurs when aneuploidy is demonstrated as an allelic imbalance at the molecular level (e.g., when one parental allele is lost in normal cells) [6]. The allelic imbalance may affect cellcycle checkpoints and centrosome numbers or may increase the frequency of structural chromosomal changes secondary to chromosome breakage and fusion [7,8]. LOH analysis has provided the opportunity to determine possible locations of critical tumor-suppressor genes and to identify possible cancer biomarkers [8,9]. In LOH studies using a panel of microsatellite markers located on different chromosomes, several researchers have identified multiple regions that are deleted in borderline ovarian tumors and invasive epithelial ovarian cancers [9,10]. SNPs are variations in the DNA sequence in a population at a single nucleotide in the human genome. So far, more than 5 million SNP loci have been identified and validated in a study population. SNPs can occur in the coding and noncoding regions of the genome, which may affect subsequent protein expression. Specific SNPs may also predispose an individual towards tumorigenesis or affect drug metabolism and 28

effect. SNP arrays based on the database of known, validated SNP loci have been developed to interrogate the genotype at 500,000 SNP loci, yielding a resolution more than a 1000-times greater than that achieved using microsatellite markers. This use of SNP arrays has led to the high-resolution mapping and detection of SNP loci exhibiting LOH and copy-number changes in ovarian cancer samples [11].

Women's Health (2008) 4(1)

CGH detects gene copy number in the DNA content of tumor cells. This allows for the analysis of gains and losses in the DNA copy sequence, gains potentially signifying the overexpression of tumor-promoting oncogenes and losses potentially signifying the underexpression of tumor-suppressor genes. The CGH method involves the competitive hybridization of tumor DNA and normal DNA, both labeled with distinct fluorescent molecules, to normal human metaphase chromosome preparations. The relative intensities of both fluorescent colors are compared using epifluorescence microscopy or quantitative image analysis, and chromosomal regions with a change in gene copy number are then identified. Past research utilizing chromosomal CGH has demonstrated that copynumber abnormalities (CNAs) are significantly more frequent in high-grade ovarian cancers than in low-grade tumors [12]. Examples include the under-representation of 11p and 13q and overrepresentation of 8q and 7p in high-grade tumors and the under-representation of 12p and over-representation of 18p in well-differentiated and moderately differentiated tumors [12]. Correlations between disease stage and CNA frequency and between chemoresistance and CNA frequency have also been evaluated [13,14]. Transcription profiling

Gene-expression profiling is an array-based technique that allows the simultaneous comparison of the expression patterns of thousands of genes and the tissue in which each gene is expressed [15–17]. There are many transcription-profiling methods, including cDNA and oligonucleotide arrays. In cDNA microarrays, the DNAs of targeted genes are imprinted onto coated glass microscope slides [18]. Total RNA from test and reference samples are fluorescently labeled by reverse transcription and hybridized to the clones on the arrays. Data from each hybridization experiment is then viewed as a normalized future science group

Biomarker discovery in ovarian cancer – REVIEW

ratio between the test and reference samples for each of the genes. Oligonucleotide arrays are similar to cDNA microarrays but are synthesized on the basis of sequence information [19]. This eliminates the need for the clones, PCR products and cDNA required to design cDNA microarrays. Oligonucleotide arrays are also designed to include a large set of probes so as to monitor the expression levels of as many genes as possible. Several researchers have reported the results of microarray analysis of ovarian cancer specimens [20–24]. Potential serum ovarian cancer biomarkers identified using a cDNA microarray system include prostasin and osteopontin, as well as an autoantibody against an epithelial cell antigen (Ep-CAM) [25–27]. Meinhold-Heerlein et al. developed an algorithm to identify secreted proteins encoded on commercial oligonucleotide arrays and applied it to gene-expression profiles of 67 ovarian cancer specimens and nine normal ovarian tissues samples [28]. They discovered that the combination of CA125 with osteopontin or kallikrein-10/matrix metalloproteinase-7 yielded a sensitivity and specificity of 96–98.7 and 99.7–100%, respectively. Expression profiling of gynecologic malignancies of different histological types and organs of origin has revealed that organ and histology contribute equally to gene expression [29,30]. Other profiling studies have revealed transcription profiles that can determine patient prognosis and survival [31,32]. Proteomic technology The proteome is the entire protein complement expressed by a genome, cell, tissue or organism. Seeking new biomarkers via analysis of the whole proteome is difficult, since proteomes differ between cells and can be constantly altered by genomic and environmental interactions. Owing to the many post-translational modifications that occur within it, the cancer proteome is estimated to include over 1.5 million proteins [33]. Initially, researchers used 2D polyacrylamide gel electrophoresis (PAGE) to probe proteomes for biomarkers, but the technique was time consuming and provided limited information regarding the proteome [34]. Newer broad-spectrum proteomic approaches – mass spectroscopy (MS), NMR spectroscopy and x-ray diffraction – have emerged and are now being incorporated into the proteomic biomarker-discovery process. MS is used to measure and analyze the mass and charge of proteins [35], whereas NMR spectroscopy and x-ray diffraction are used to evaluate the 3D structure of proteins. future science group

www.futuremedicine.com

These newer proteomic techniques help identify the components of the cancer proteome and allow comparison of protein expression in normal versus malignant tissues in various stages of disease and in response to therapeutic intervention. The structural knowledge of proteins gained through NMR spectroscopy and x-ray diffraction analysis can illustrate how the proteins interact with other molecules, catalyze enzymatic reactions and are affected by oncogenic mutations. In addition, they allow insights to be made into how proteins are post-translationally modified and the effect that these alterations have on protein function. Gel-based proteomics

2D-PAGE has been the foundation of proteomic biomarker identification, being used primarily to identify and compare protein expression in normal versus tumor tissues [36]. Over 1000 proteins can be resolved per gel and separated on the basis of charge and size [37]. The desired proteins can then be excised, purified and identified. However, as stated previously, the 2D-PAGE method is extremely time consuming and limited. All potential biomarkers must be validated and tested prior to clinical application, and the method’s sensitivity is severely impaired by its inability to detect low-abundance proteins, hydrophobic proteins and proteins of less than 15 kDa or more than 150 kDa [36,38]. Mass spectroscopy

MS allows the analysis of thousands of complex proteins on a variety of platforms. The technique depends on the ionization of large biomolecules and identifies substances by sorting them according to their mass:charge (m/z) ratios [37]. A protein is first digested with an enzyme, usually trypsin, at specific amino acid sequences. The peptides are energized, usually by either electrospray ionization or laser, and then separated in the spectrometer using a variety of methods depending on the type of spectrometer. The separated peptides are deflected onto a detector that measures the signal intensity of each fragment and the m/z ratios of all peptide ions produced [37]. A mass spectrum is created by graphing the signal intensity, or relative fragment abundance, against the m/z ratio. The peptides are then further fragmented into a second mass spectrum, on which each peptide fragment differs by one amino acid, in order to determine the peptide sequence [37]. The sequence is then used to probe protein databases in order to identify the original protein. 29

REVIEW – Tung, Wong & Mok

The use of laser to ionize peptides can involve either SELDI-TOF or MALDI-TOF, by which a laser is directed at a protein-binding chip containing many patient samples. This interaction causes adherent proteins to be desorbed and launched as protonated and charged ions. The time of flight of the ionized peptides depends on the m/z ratio and is determined prior to hitting the detector. Again, the detector plate records the signal intensity at a certain m/z ratio to generate a mass spectrum. SELDI-TOF is considered to have the higher throughput of the two laser technologies. Vast amounts of data have been produced from a small amount of starting material and analyzed within a very short period of time [36,39]. MS continues to play a critical role in the identification of new ovarian cancer biomarkers. Elevated serum levels of haptoglobin-α chain and inter-α-trypsin inhibitor heavy-chain H4 have been identified in ovarian cancer patients using this technology [40,41]. Important considerations in using proteomic technology

Use of the proteomic techniques to identify new ovarian cancer biomarkers requires careful planning and careful selection of technologies to be used. An important consideration is sample collection. Serum and plasma are now the most widely used bodily fluids, but urine or saliva have also been used. Another important consideration is the fact that validation of newly discovered biomarkers remains a challenge. ‘Between-laboratory’ and ‘within-laboratory’ reproducibility has been demonstrated recently using SELDI-TOF-MS serum profiling [42]. Validation studies need to ensure the specificity and reproducibility of the biomarkers and should address experiment design and appropriate controls [33]. Specifically, areas such as specimen collection, specimen handling and bioinformatic data analysis require further study and examination. Bioinformatics

Proteomic pattern diagnostics is a bioinformatics-based means of searching for a discriminating series of proteins or protein patterns that, when evaluated together, help to distinguish between unaffected and affected individuals. In a landmark report, Petricoin et al. described the first pattern-recognition algorithms and their application to ovarian cancer detection [43]. In brief, they generated proteomic spectra using 30

Women's Health (2008) 4(1)

SELDI-TOF, and analyzed serum samples from 50 unaffected individuals and 50 ovarian cancer patients to create a training set of mass spectra. They then applied the resulting diagnostic pattern algorithm to 116 masked samples, thereby yielding 100% sensitivity, 95% specificity and 94% PPV in identifying patients with malignant and benign disease. The bioinformatics approach utilized by Petricoin et al. was a genetic algorithm using artificial-intelligence computerbased models to identify the protein spectra by unsupervised testing and retesting. Other bioinformatics models have also been used to identify novel biomarkers. Zhu et al. created an algorithm in which all biomarkers whose expression levels differed significantly between affected and unaffected individuals were selected from mass spectra using the random-field theory [44]. The best discriminating pattern was then chosen using the best-subset discriminant analysis method. This method yielded 100% sensitivity and 100% specificity. Logical analysis of data has also been applied to create analytical tests that yield 100% sensitivity and 100% specificity [45]. The difficulty in these approaches is due to the complexity of the serum proteome and peptidome, and as a result, still requires vigorous clinical validation. Many identified potential blood peptide biomarker levels are constantly in flux owing to normal physiological and various nondisease-related factors. Therefore, to minimize bias, specific parameters must be implemented during bioinformatics biomarker discovery [46]. The use of inflammatory controls and benign disease as part of discovery sets is crucial to help minimize fluctuations, and these controls must be matched in every possible epidemiological and physiological parameter. The handling and storage of all specimens must occur in a standardized and rapid manner to prevent further bias. Furthermore, the identification and validation of each candidate biomarker in the panel is extremely labor intensive, requiring repetitive iterations and validations to ensure specificity. Metabolomics

Cellular metabolites are the final products of the interactions between gene expression, protein expression and the cellular environment. The complex biochemical network of these lipids, small peptides, vitamins, protein cofactors and other metabolic substrates and products that interact with each other and with other biological future science group

Biomarker discovery in ovarian cancer – REVIEW

macromolecules is called the ‘metabolome’ [47]. The concentration and ultimate identities of the many components of a metabolome rest on their genomic and proteomic interactions within the tissues. These metabolites are involved in cell transformation from normal to malignant and are present in detectable levels in body fluids [48]. Unfortunately, using this approach in the search for ovarian cancer biomarkers is severely hampered by the lack of a single analytical platform to measure the metabolites and by the large number of unknown metabolites with dynamic ranges in human serum that must be determined in order to complete metabolite profiling [49]. However, steps are being taken to overcome these obstacles. In January 2007, the Human Metabolome Project completed the first draft of the human metabolome, identifying 2500 metabolites, 1200 drugs and 3500 food components in the human body [50]. Metabolomic analysis involves two steps and uses techniques similar to those used for proteomic analysis. In the first step, global analysis via NMR spectroscopy or MS is used to identify and quantify all possible metabolites in a given biological matrix [51]. Interpretation of the complex datasets generated by these studies, which usually requires visualization software and chemometric and bioinformatic methods [52]. In the second step, the substances associated with these fingerprints are identified and then combined to define a biological or clinical end point [52]. Clinical validation The final and crucial step prior to clinical biomarker application is validation of the potential biomarkers. The measurement platform is tested for sensitivity and precision, and once proven to be reliable, the test is then introduced into independent clinical studies [46]. The sample size of these studies depends on the level of statistical power desired. This relies on both the performance of the peptide analyte panel in the platform-validation discovery phase and the clinical use of the biomarker [46]. For example, general population screening for ovarian cancer would require a larger sample size for clinical validation compared with biomarkers intended for recurrence or treatment response. The true specificity, sensitivity and positive predictive values of candidate biomarkers are elicited through these clinical validation studies since these do not translate from experimental test populations [46]. future science group

www.futuremedicine.com

Uses for biomarkers Diagnosis of early ovarian cancer The identification of a novel biomarker of early ovarian cancer would dramatically improve the ultimate prognosis for these patients. Several different techniques, including pelvic ultrasonography, serum CA125 screening and a combination of the two, have been evaluated as possible candidates [53–60]. However, to date, none of these modalities has achieved the sensitivity, specificity and PPV crucially important for any screening test. Pelvic ultrasonography has excellent sensitivity for the detection of ovarian lesions. However, its poor specificity and poor diagnostic reproducibility prohibit its use as a primary screening tool [53,55,58]. Serum CA125 screening is useful because serum CA125 levels are elevated in 50% of stage I and 90% of stage II ovarian cancers [57]. Nonetheless, this screening tool’s false-positive rate is high because there are many benign gynecologic and nongynecologic conditions that can also result in serum CA125 elevation [54]. The new ‘risk of ovarian cancer’ screening method uses an algorithm that incorporates patient age, absolute CA125 levels and rate of change of CA125 levels. This method has demonstrated 99.8% specificity and 19% PPV for detecting primary invasive ovarian cancer [60]. One of the more important aspects of this method is the rate of change in the levels of the biomarkers: elevation and then decline may indicate a transient condition; stable elevation may indicate a chronic, benign condition; elevation and then a further increase may indicate ovarian cancer [60,61]. Currently, two large, ongoing randomized multicenter trials are evaluating a multimodal screening method for ovarian cancer. Preliminary data from the Prostate, Lung, Colorectal and Ovarian Cancer Screening trial shows that an abnormal serum CA125 test has a 3.7% PPV for invasive cancer, an abnormal transvaginal ultrasound has a 1.0% PPV, and the combination of both has a 23.5% PPV [59]. Further studies are being conducted to determine the efficacy of this approach in lowering ovarian cancer mortality. The other screening trial is the UK Collaborative Trial of Ovarian Cancer Screening, which has recruited 200,000 postmenopausal women aged between 50 and 74 years and randomized them to a control group or annual serum CA125 or transvaginal ultrasound screening based on the risk-of-ovarian-cancer algorithm, with the objective of 31

REVIEW – Tung, Wong & Mok

lowering mortality by 30% [202]. As approximately 20% of ovarian cancers express little or no CA125, the use of multiple serum biomarkers in conjunction with these screening methods has been postulated to increase sensitivity and specificity of ovarian cancer screening. Ovarian cancer screening Ovarian cancer is an extremely heterogenous disease and thus poses a large challenge to biomarker discovery. There are various histological subtypes of epithelial ovarian cancer (i.e., serous, endometrioid and clear-cell carcinoma), and each is marked by different molecular characteristics and features. As a result, detecting ovarian cancer in its early stages will require a panel of tumor markers. The most comprehensive list of clinically relevant epithelial ovarian cancer biomarkers identified to date is found in Table 1. One study compared the gene expression profiles of 42 ovarian cancers of different histological subtypes with those of five pools of normal ovarian epithelial tissue [62]. Recursive descent partition analysis using one panel of potential tumor markers (i.e., HE4, CA125 and MUC1) could distinguish between tumor and normal ovarian tissues; immunohistochemical analysis using another panel (i.e., CLDN3, CA125, MUC1 and VEGF) could detect all tumors. Another study identified a multivariate model in which a panel of biomarkers, including CA125, apolipoprotein A1, transthyretin and inter-α-trypsin inhibitor heavy-chain H4, had a sensitivity for detecting early-stage ovarian cancer of 74 versus 65% for CA125 alone at a matched specificity of 97% [63]. In yet another study, a combination of CA125-II, CA15–3, CA72–4 and macrophage colony-stimulating factor had a specificity for detecting early-stage disease of 75 versus 48% for CA125-II alone at a fixed specificity of 98% [64]. Mesothelin, a cell-surface protein, has been identified as a potential early diagnostic biomarker in both serum and urine [65,66]. Lysophosphatidic acid, a lipid overproduced in the serum and ascitic fluid of ovarian cancer patients, appears to be a useful diagnostic and prognostic marker [67,68]. Other useful markers, identified in immunohistochemical studies, include kallikrein 10, kallikrein 6, osteopontin and claudin 3 (expressed in 100% of all non-CA125-expressing ovarian cancer specimens), and DF3, VEGF, MUC1, mesothelin, HE4 and CA19–9 (expressed in 29–95% of such specimens) [69]. Of all these potential 32

Women's Health (2008) 4(1)

markers, HE4 is most like CA125 in terms of sensitivity for late-stage disease; in addition, HE4 also appears to have greater specificity for ovarian cancer than for normal tissue and benign disease [69,70]. Treatment response evaluation Tumor markers are useful for monitoring a patient’s response to surgery, chemotherapy or radiation therapy. Often after tumor-reductive surgery, residual disease in the form of small tumor nodules remains and cannot be detected by traditional radiologic methods (e.g., CT, MRI or PET). As a result, serum markers such as CA125 are extremely helpful in determining whether a patient is responding appropriately to treatment. A rising serum CA125 level correlates with disease progression in approximately 90% of cases, and persistently elevated CA125 levels correlate with persistent disease in 95% of cases at the time of second-look laparotomy [71]. Furthermore, patients whose tumors are in remission but ultimately recur often have an elevated CA125 level prior to any other evidence of disease recurrence [71]. However, this is not true for all patients with recurrent disease, and some patients who have normal CA125 levels after treatment may still have persistent small-volume disease. In addition, no studies have demonstrated that early treatment in response to a rising CA125 level in the absence of symptoms improves patient survival [72]. Expression of tumor markers could be useful end points in clinical trial design. For example, investigators could use the levels of serum CA125 to determine whether patients are responding to a particular study treatment. In 19 Phase II trials of 14 different cytotoxic drugs, a 50–75% decrease in CA125 level correlated with response rate [73]. At present, approximately 70% of ovarian cancer patients are sensitive to platinum-based chemotherapeutics, whether administered with or without a taxane drug [71]. New potential biomarkers of platinum resistance include p53, ERCC1, copper transporters and XIAP [74–77]. Several biomarkers have also been identified for taxane resistance, such as MDR1 and tubulin mutations [78,79]. Other potential genetic predictors of sensitivity to platinum-based drugs are MYO5C, SPINK1, ARMCX3, PLEK2 and PRSS11, and for taxanes, TNFSF13B, IFIT3 and BTN3A2 [80]. Further research is needed to determine the clinical applicability of these new gene markers. future science group

Biomarker discovery in ovarian cancer – REVIEW

Table 1. Novel potential ovarian cancer biomarkers . Name

Use

Type

Description

Apolipoprotein A1

Screening

Protein

Major apolipoprotein of high-density lipoprotein

Ref.

B7-H4

Screening

Protein

Cell-surface protein, unknown function but possibly involved with malignant transformation of epithelial cells

[95,96]

Bcl-2

Screening

Gene/protein

Involved in inhibition of apoptosis

[97,98]

CA15–3

Screening

Protein

Carbohydrate antigen, tumor marker for breast cancer

[64,99]

CA19–9

Screening

Protein

Tumor marker for pancreatic cancer and coexpressed with CA125 in ovarian cancer

CA72–4

Screening

Protein

Antibody assay that reacts with TAG-72 antigen

CASA

Treatment response

Protein

High-molecular-weight mucin glycoprotein

[91,101,102]

CLDN3

Screening

Protein

Involved with epithelial cell tight junctions, critical to the maintenance of cell polarity and permeability

[69,103,104]

Copper transporters (ATP7A, ATP7B, CTR1)

Treatment response to platinum-based drugs, prognosis

Protein

Copper efflux transporter

[76,105,106]

DcR3

Screening

Protein

TNF receptor, may inhibit FasL-mediated apoptosis

[95]

DF3

Screening

Co-expressed with CA125

[69]

DDR1

Screening

Protein

Tyrosine receptor kinase activated by collagen, involved in cell-matrix communication

EphA2

Prognosis

Protein

Transmembrane protein involved with angiogenesis and tumor invasion

Ep-CAM

Screening

Protein

Member of cell-adhesion molecule family

ERCC1

Treatment response

Gene

Involved in the nucleotide excision repair pathways

FGF-1, bFGF

Prognosis, treatment response to paclitaxel

Protein

Growth factor involved in cell growth and differentiation

HER-2/neu

Screening

Gene

Proto-oncogene

HE4

Screening

Protein

Glycoprotein member of a family of 4-disulfide core proteins, unknown function

[69,70,103]

HIF-α

Prognosis

Gene

α/β heterodimeric DNA-binding complex that directs

[112]

[63,93,94]

[69]

[64,100]

[104]

[107,108]

[104] [75,109] [110,111]

[86,87,99]

extensive transcriptional response involving the induction of genes relevant to tumor progression IDO

Prognosis, treatment response to paclitaxel

Protein

Tryptophan-catabolizing enzyme that induces immune tolerance

ITIH4

Screening

Protein

Acute-phase protein

Kallikreins (KLK5, 6, 7, 10, 11 and 14)

Screening, treatment decisions, prognosis

Protein

Family of 15 secreted serine proteases

LPA

Screening, prognosis

Lipid

Phospholipid associated with apoptosis suppression of tumor cells

M-CSF, CSF-1

Prognosis

Protein

Hematopoietic cytokine controlling macrophage and monocytic progenitor proliferation

MMP-2, MMP-7, MMP-9

Screening, prognosis

Protein

Involved in degradation of extracellular matrix

MEKK3

Treatment response

Gene

Serine/threonine protein kinase, upstream regulators of the three-module cascade of MAPK pathway

[113,114]

[63]

[67,68]

[64,82–84,92]

[28,115,116]

[117]

API: α-1-proteinase inhibitor; CASA: Cancer associated serum antigen; CLDN3: Claudin 3; DDR1: Discoidin domain receptor 1; Ep-CAM: Epithelial cell-adhesion molecule; ERCC1: Excision repair cross-complementation group 1; HE4: Human epididymis protein; HIF: Hypoxia-inducible factor; IDO: Indoleamine-2,3-dioxygenase; ITIH4: Inter-α-trypsin inhibitor heavy chain H4; LPA: Lysophosphatidic acid; M-CSF: Macrophage colony-stimulating factor; MDR: Multidrug resistance; MMP: Matrix metalloproteinase; PAI: Plasminogen activator inhibitor; sEGFR: p110 soluble epidermal growth factor receptor; SLPI: Secretory leukoprotease inhibitor; TATI: Tumor-associated trypsin inhibitor; THBS: Thrombospondin.

future science group

www.futuremedicine.com

33

REVIEW – Tung, Wong & Mok

Table 1. Novel potential ovarian cancer biomarkers (cont.). Name

Use

Type

Description

Mesothelin (serum and urine)

Screening

Protein

Glycosylphosphatidylinositol-anchored cell-surface glycoprotein

Ref.

MUC1

Screening

Protein

Glycosylated type I transmembrane glycoprotein

MDR1

Treatment response

Gene

Encodes a glycoprotein that functions as a transmembrane drug-efflux pump

Neu3

Prognosis

Protein

Plasma membrane-associated sialidase

nm23

Screening

Gene/protein

Protein expressed in non-metastatic cells

Osteopontin

Screening

Protein

Adhesive glycoprotein

OVX1

Screening

Protein

Modified Lewis X determinant on mucin

sEGFR

Screening

Protein

Glycoprotein

p53

Treatment response, prognosis

Gene

Tumor-suppressor gene

PAI-1

Prognosis

Protein

Involved in proteolysis

Prostasin

Screening

Protein

Serine protease, unknown function

SLPI

Screening

Protein

Serine protease inhibitor, member of a family of 4-disulfide core proteins

Spondin 2

Screening

Protein

Unknown function

Survivin

Prognosis

Protein

Caspase inhibitor-of-apoptosis protein

Tetranectin

Treatment response, prognosis

Protein

Glycoprotein involved in tissue remodeling

THBS-1

Prognosis

Protein

Involved with tumor-associated angiogenesis

Total inhibin

Screening

Protein

Ovary-secreted glycoprotein

Transthyretin

Screening

Protein

Binding protein to transport thyroxine and vitamin A

Trophonin

Treatment response to cisplatin, prognosis

Protein

Homophilic adhesion molecule involved in blastocyst implantation

Trypsin-2-API

Prognosis

Protein

Proteinase inhibitor

Tubulin mutations

Treatment response to paclitaxel

Protein

Point mutations in tubulin at paclitaxel binding site

Tumor M2-PK

Prognosis

Protein

Pyruvate kinase isoenzyme

[131]

TATI

Screening, prognosis

Protein

Trypsin inhibitor

[129]

Urine cyclic GMP

Prognosis

Protein

Cyclic nucleotide that acts as second messenger to activated intracellular protein kinases

[132]

Urokinase-type plasminogen activator

Prognosis

Protein

Involved in proteolysis

[125]

VEGF

Prognosis

Protein

Involved with angiogenesis and vascular permeability

XIAP

Treatment response, prognosis

Protein

Caspase inhibitor of apoptosis protein

YKL-40

Screening, treatment response

Protein

Secreted glycoprotein in chitinase protein family, unknown function

[65,66,69]

[69,103,118] [78]

[119] [99,120] [25,28,69, 121,122] [91,92] [123] [74,98,99,124]

[125] [27,126] [28,90]

[95] [85] [91,101]

[124] [127] [63,93,94] [128]

[129] [79,130]

[69,133]

[77,85]

[101,134,135]

API: α-1-proteinase inhibitor; CASA: Cancer associated serum antigen; CLDN3: Claudin 3; DDR1: Discoidin domain receptor 1; Ep-CAM: Epithelial cell-adhesion molecule; ERCC1: Excision repair cross-complementation group 1; HE4: Human epididymis protein; HIF: Hypoxia-inducible factor; IDO: Indoleamine-2,3-dioxygenase; ITIH4: Inter-α-trypsin inhibitor heavy chain H4; LPA: Lysophosphatidic acid; M-CSF: Macrophage colony-stimulating factor; MDR: Multidrug resistance; MMP: Matrix metalloproteinase; PAI: Plasminogen activator inhibitor; sEGFR: p110 soluble epidermal growth factor receptor; SLPI: Secretory leukoprotease inhibitor; TATI: Tumor-associated trypsin inhibitor; THBS: Thrombospondin.

Prognosis Biomarkers can also be utilized to help predict a patient’s ultimate prognosis. CA125 half-life appears to correlate with prognosis [81]. For 34

Women's Health (2008) 4(1)

example, the CA125 half-life in patients whose disease is completely resected is 6–14 days; in those with residual disease, the half-life is longer [71,72]. A response to chemotherapy treatment future science group

Biomarker discovery in ovarian cancer – REVIEW

will also shorten the CA125 half-life. Poor prognostic factors include a half-life of greater than 20 days during chemotherapy and failure of CA125 levels to normalize after three cycles of chemotherapy. The macrophage colony-stimulating factor CSF-1 has been shown to correlate with ovarian cancer cell invasion and to be associated with poor prognosis when present at elevated levels in serum and ascitic fluid [82–84]. Other already identified biomarkers, such as nuclear overexpression of survivin, can indicate improved outcome of ovarian cancer patients [85]. Meanwhile, the prognostic value of other markers (e.g., Her2/neu) remains controversial and will require validation via further research and investigation [86,87]. Conclusion The complexity of ovarian cancer has made the discovery of novel ovarian cancer biomarkers a challenging and rigorous task. Nonetheless, such tumor markers are important for their potential uses in early disease detection and screening, treatment-response evaluation and planning, and prognosis. New technologies and methods have dramatically changed the way that these biomarkers are sought out. Genomic technologies now allow gene expression, association, regulation and function to be systematically evaluated for possible involvement in tumorigenesis. Proteomic studies of protein expression, structure and function have revealed the complexity of the human proteome due to posttranslational modifications and molecular interactions. By combining genomic and proteomic data, metabolomics has opened a new window into the complex biochemical network of macromolecules and metabolites and their possible roles in ovarian cancer. The prognosis for ovarian cancer is good, provided tumors are found early. Unfortunately, most ovarian tumors are only diagnosed after they have become well advanced, mainly because the symptoms of early disease are vague, and as a result have a much poorer prognosis. Therefore, the search for early screening tools is urgent. The best-studied ovarian cancer biomarkers to date – serum CA125, pelvic ultrasound and combination of the two – still lack the specificity, sensitivity and PPV necessary for clinical implementation. Furthermore, the heterogeneity of ovarian cancer in its various forms, stages and histopathologic subtypes prevents the use of a solitary tumor marker and, in future science group

www.futuremedicine.com

fact, necessitates the use of a panel of biomarkers. Biomarkers such as inhibin A, LDH, AFP and β-HCG are currently used to distinguish and follow nonepithelial ovarian neoplasms, including granulosa cell and other sexcord/stromal cell tumors and ovarian germ cell tumors. However, many of these patients do not express elevated levels of these biomarkers, and despite the rarity of these diseases, more-specific tumor markers are needed to monitor disease incidence and progression [88,89]. Many new biomarkers have been identified for epithelial and nonepithelial ovarian cancers, and several trials are now assessing their clinical relevance when combined with serum CA125 for the early diagnosis of the disease. Fortunately, the list continues to expand as new genes and proteins are discovered and assessed for their potential as diagnostic and prognostic biomarkers. Future perspective Advances in genomic technologies have led to the discovery of many promising biomarkers for use in screening and predicting the treatment response and outcomes of ovarian cancer. On the horizon are emerging technologies (e.g., medical resequencing) that will further enhance the ability to predict cancer risk and drug sensitivity. However, because a positive test could lead to surgery, the PPV of any proposed panel of biomarkers would need to be as high as possible to minimize the risk of unnecessary surgery associated with a false-positive result. As greater numbers of tumor samples are screened against such panels, the power to validate their utility will increase, along with the chances that novel molecular targets will be identified and less toxic therapies will be developed. It is hoped that, by revealing the genetic origins of ovarian cancer, the findings of genomic, proteomic, bioinformatic and metabolomic studies will translate into earlier diagnosis and better patient outcomes. Despite some successes, much more research and collaborative effort are needed to identify the proper combination of ovarian cancer biomarkers required for early detection and to resolve conflicting data regarding the utility of several of the newer biomarkers (e.g., secretory leukoprotease inhibitor [28,90] and OVX1 [91,92]). In addition, the sensitivities of all candidate biomarkers need to be further evaluated, since all previous estimates are based on the analysis of preoperative samples obtained from patients who had clinically detectable early- or late-stage disease. The best hope of all is for a simple blood 35

REVIEW – Tung, Wong & Mok

test for early-stage (and therefore highly curable) ovarian cancer that would be available in the near future. Financial & competing interests disclosure The authors have no relevant affiliations or financial involvement with any organization or entity with a

financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

Executive summary Screening biomarker criteria • Novel diagnostic biomarkers should be noninvasive and inexpensive and have a high sensitivity for early-stage disease, with a high specificity and positive predictive value. • Any new ovarian cancer-screening strategy should have a minimum positive predictive value of 10% and a minimum specificity of 99.6%. Higher risk populations (e.g., individuals with a strong family history of ovarian or breast cancer) may be screened using a method of lower specificity. Tools for identifying ovarian cancer biomarkers • Biomarkers are being identified using new technologies in the fields of genomics, proteomics, bioinformatics and metabolomics. • Genomics allows the discovery of novel gene biomarkers through the systematic evaluation of gene information and association via loss of heterozygosity analysis, comparative genomic hybridization and transcription profiling. • Proteomics allows the discovery of novel biomarkers through the analysis of protein structure, function and composition via mass spectroscopy and other technologies. Bioinformatics allow affected and unaffected patients to be distinguished through the identification of distinct protein series or patterns. • Metabolomics allows the discovery of potential biomarkers through the evaluation of macromolecules and cell metabolites. • Clinical validation of all new potential biomarkers is crucial to ensure sensitivity and precision of the measurement platform. Uses for biomarkers: diagnosis of early cancer • Pelvic ultrasonography has a high sensitivity for ovarian masses but has low specificity and diagnostic reproducibility in ovarian cancer detection. • Serum CA125 is elevated in 50% of stage I and 90% of stage II ovarian cancers but can be falsely positive in patients with benign disease. • Large, randomized multicenter trials, including the Prostate, Lung, Colorectal, and Ovarian Cancer Screening trial and the UK Collaborative Trial of Ovarian Cancer Screening, are evaluating potential multimodal screening methods for ovarian cancer using a combination of CA125 levels, transvaginal ultrasound and ‘risk of ovarian cancer’ screening tools. Uses for biomarkers: ovarian cancer screening • The heterogeneity of ovarian cancer will likely require the use of a panel of biomarkers for screening early-stage disease. • Several studies have been performed that indicate that a combination of CA125 with other biomarkers, such as CA15–3, CA72–4 and apolipoprotein A1, increases the sensitivity and specificity of detecting early-stage disease compared with CA125 alone. Uses for biomarkers: treatment response evaluation • Elevated CA125 levels are often the first sign of disease recurrence prior to any other evidence of tumor burden. • Biomarkers are useful to monitor disease response to treatment since they often measure tumor burden, which cannot be detected by traditional radiologic modalities. • Several new potential biomarkers have been identified to predict tumor sensitivity to various chemotherapeutic agents, including taxanes and platinum-based drugs. Uses for biomarkers: prognosis • Biomarkers can be used to correlate and predict patient prognosis. Increased half-life of CA125 levels and macrophage colony-stimulating factor appear to correlate with poor prognosis. • Clinical validation of other biomarkers, including survivin and Her2/neu, are needed to determine their clinical applicability to indicate patient prognosis. Future perspective • New emerging techniques (e.g., medical resequencing) will further aid in predicting cancer risk and disease sensitivity to various chemotherapeutic agents. • More research and collaborative efforts are needed to identify the proper combination of ovarian cancer biomarkers and to resolve conflicting data regarding some of the newly identified biomarkers.

36

Women's Health (2008) 4(1)

future science group

Biomarker discovery in ovarian cancer – REVIEW

Bibliography Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers. 1. Hoskins WJ: Prospective on ovarian cancer: why prevent? J. Cell Biochem. Suppl. 23, 189–199 (1995). 2. Biomarkers Definitions Working Group: Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 69(3), 89–95 (2001). 3. Bast RC Jr, Urban N, Shridhar V et al.: Early detection of ovarian cancer: promise and reality. Cancer Treat. Res. 107, 61–97 (2002). 4. Jacobs IJ, Menon U: Progress and challenges in screening for early detection of ovarian cancer. Mol. Cell Proteomics 3(4), 355–366 (2004). 5. Ye B, Gagnon A, Mok SC: Recent technical strategies to identify diagnostic biomarkers for ovarian cancer. Expert Rev. Proteomics 4(1), 121–131 (2007). 6. Cavenee WK, Dryja TP, Phillips RA et al.: Expression of recessive alleles by chromosomal mechanisms in retinoblastoma. Nature 305(5937), 779–784 (1983). •• Landmark paper describing loss of heterozygosity causing a predisposition for cancer. 7. Doxsey S: The centrosome – a tiny organelle with big potential. Nat. Genet. 20(2), 104–106 (1998). 8. Thiagalingam S, Laken S, Willson JK et al.: Mechanisms underlying losses of heterozygosity in human colorectal cancers. Proc. Natl Acad. Sci. USA 98(5), 2698–702 (2001). • Classic paper describing a new model of aneuploidy in cancer. 9. Liu Y, Ganesan TS: Tumour suppressor genes in sporadic epithelial ovarian cancer. Reproduction 123(3), 341–353 (2002). 10. Auersperg N, Edelson MI, Mok SC, Johnson SW, Hamilton TC: The biology of ovarian cancer. Semin. Oncol. 25(3), 281–304 (1998). 11. Gorringe KL, Jacobs S, Thompson ER et al.: High-resolution single nucleotide polymorphism array analysis of epithelial ovarian cancer reveals numerous microdeletions and amplifications. Clin. Cancer Res. 13(16), 4731–4739 (2007). 12. Kommoss F, Wolfle J, Bauknecht T et al.: Co-expression of M-CSF transcripts and protein, FMS (M-CSF receptor) transcripts and protein, and steroid receptor content in adenocarcinomas of the ovary. J. Pathol. 174(2), 111–119 (1994).

future science group

13.

14.

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

Takahashi K, Yoshida H, Sonoda S et al.: Expression of a rat ovary-independent mammary tumor-associated antigen defined by a monoclonal antibody, TAK-B1. Jpn J. Cancer Res. 82(5), 577–584 (1991). Wolf NG, Abdul-Karim FW, Farver C et al.: Analysis of ovarian borderline tumors using comparative genomic hybridization and fluorescence in situ hybridization. Genes Chromosomes Cancer 25(4), 307–315 (1999). Schena M, Shalon D, Davis RW, Brown PO: Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science 270(5235), 467–470 (1995). Schena M, Shalon D, Heller R et al.: Parallel human genome analysis: microarray-based expression monitoring of 1000 genes. Proc. Natl Acad. Sci. USA 93(20), 10614–10619 (1996). Lockhart DJ, Dong H, Byrne MC et al.: Expression monitoring by hybridization to high-density oligonucleotide arrays. Nat. Biotechnol. 14(13), 1675–1680 (1996). Duggan DJ, Bittner M, Chen Y, Meltzer P, Trent JM: Expression profiling using cDNA microarrays. Nat. Genet. 21(Suppl. 1), 10–14 (1999). Lipshutz RJ, Fodor SP, Gingeras TR, Lockhart DJ: High density synthetic oligonucleotide arrays. Nat. Genet. 21(Suppl. 1), 20–24 (1999). Ismail RS, Baldwin RL, Fang J et al.: Differential gene expression between normal and tumor-derived ovarian epithelial cells. Cancer Res. 60(23), 6744–6749 (2000). Wong KK, Cheng RS, Mok SC: Identification of differentially expressed genes from ovarian cancer cells by MICROMAX cDNA microarray system. Biotechniques 30(3), 670–675 (2001). Matei D, Graeber TG, Baldwin RL et al.: Gene expression in epithelial ovarian carcinoma. Oncogene 21(41), 6289–6298 (2002). Manderson EN, Mes-Masson AM, Novak J et al.: Expression profiles of 290 ESTs mapped to chromosome 3 in human epithelial ovarian cancer cell lines using DNA expression oligonucleotide microarrays. Genome Res. 12(1), 112–121 (2002). Shridhar V, Lee J, Pandita A et al.: Genetic analysis of early- versus late-stage ovarian tumors. Cancer Res. 61(15), 5895–5904 (2001). Kim JH, Skates SJ, Uede T et al.: Osteopontin as a potential diagnostic biomarker for ovarian cancer. JAMA 287(13), 1671–1679 (2002).

www.futuremedicine.com

26.

27.

28.

29.

30.

31.

32. 33.

34.

35.

36.

37.

38.

Kim JH, Herlyn D, Wong KK et al.: Identification of epithelial cell adhesion molecule autoantibody in patients with ovarian cancer. Clin. Cancer Res. 9(13), 4782–4791 (2003). Mok SC, Chao J, Skates S et al.: Prostasin, a potential serum marker for ovarian cancer: identification through microarray technology. J. Natl Cancer Inst. 93(19), 1458–1464 (2001). Meinhold-Heerlein I, Bauerschlag D, Zhou Y et al.: An integrated clinical-genomics approach identifies a candidate multi-analyte blood test for serous ovarian carcinoma. Clin. Cancer Res. 13(2 Pt 1), 458–466 (2007). Shedden KA, Kshirsagar MP, Schwartz DR et al.: Histologic type, organ of origin, and Wnt pathway status: effect on gene expression in ovarian and uterine carcinomas. Clin. Cancer Res. 11(6), 2123–2131 (2005). Zorn KK, Bonome T, Gangi L et al.: Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer. Clin. Cancer Res. 11(18), 6422–6430 (2005). Spentzos D, Levine DA, Ramoni MF et al.: Gene expression signature with independent prognostic significance in epithelial ovarian cancer. J. Clin. Oncol. 22(23), 4700–4710 (2004). Berchuck A: Biomarkers in the ovary. J. Cell Biochem. 23, 223–226 (1995). Howell NR, Zheng W, Cheng L et al.: Carcinomas of ovary and lung with clear cell features: can immunohistochemistry help in differential diagnosis? Int. J. Gynecol. Pathol. 26(2), 134–140 (2007). Posadas EM, Davidson B, Kohn EC: Proteomics and ovarian cancer: implications for diagnosis and treatment: a critical review of the recent literature. Curr. Opin. Oncol. 16(5), 478–484 (2004). Kamel-Reid S, Arrowsmith CH, Reis PP, Squire JA: Methods of molecular analysis. In: The Basic Science of Oncology. Tannock IF, Hill RP, Bristow RG, Harrington L (Eds). McGraw–Hill, NY, USA, 49–76 (2005). Wulfkuhle JD, Liotta LA, Petricoin EF: Proteomic applications for the early detection of cancer. Nat. Rev. Cancer 3(4), 267–275 (2003). Kavallaris M, Marshall GM: Proteomics and disease: opportunities and challenges. Med. J. Aust. 182(11), 575–579 (2005). de Nictolis M, Garbisa S, Lucarini G et al.: 72kilodalton type IV collagenase, type IV collagen, and Ki 67 antigen in serous tumors or the ovary: a clinicopathologic, immunohistochemical, and serological study. Int. J. Gynecol. Pathol. 15(2), 102–109 (1996).

37

REVIEW – Tung, Wong & Mok

39.

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

38

Lee CJ, Ariztia EV, Fishman DA: Conventional and proteomic technologies for the detection of early stage malignancies: markers for ovarian cancer. Crit. Rev. Clin. Lab Sci. 44(1), 87–114 (2007). Ye B, Cramer DW, Skates SJ et al.: Haptoglobin-α subunit as potential serum biomarker in ovarian cancer: identification and characterization using proteomic profiling and mass spectrometry. Clin. Cancer Res. 9(8), 2904–2911 (2003). Acs G, Pasha T, Zhang PJ: WT1 is differentially expressed in serous, endometrioid, clear cell, and mucinous carcinomas of the peritoneum, fallopian tube, ovary, and endometrium. Int. J. Gynecol. Pathol. 23(2), 110–118 (2004). Semmes OJ, Feng Z, Adam BL et al.: Evaluation of serum protein profiling by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry for the detection of prostate cancer: I. Assessment of platform reproducibility. Clin. Chem. 51(1), 102–112 (2005). Petricoin EF, Ardekani AM, Hitt BA et al.: Use of proteomic patterns in serum to identify ovarian cancer. Lancet 359(9306), 572–577 (2002). Zhu W, Wang X, Ma Y et al.: Detection of cancer-specific markers amid massive mass spectral data. Proc. Natl Acad. Sci. USA 100(25), 14666–14671 (2003). Alexe G, Alexe S, Liotta LA et al.: Ovarian cancer detection by logical analysis of proteomic data. Proteomics 4(3), 766–783 (2004). Petricoin EF, Belluco C, Araujo RP, Liotta LA: The blood peptidome: a higher dimension of information content for cancer biomarker discovery. Nat. Rev. Cancer 6(12), 961–967 (2006). Beecher CWW: The human metabolome. In: Metabolic Profiling: Its Role in Biomarker Discovery and Gene Function Analysis. Harrigan GG, Goodacre R (Eds). Kluwer Academic Publishers, MA, USA 311–320 (2003). Maruvada P, Wang W, Wagner PD, Srivastava S: Biomarkers in molecular medicine: cancer detection and diagnosis. Biotechniques (Suppl.) 9–15 (2005). Vang R, Gown AM, Farinola M et al.: p16 expression in primary ovarian mucinous and endometrioid tumors and metastatic adenocarcinomas in the ovary: utility for identification of metastatic HPV-related endocervical adenocarcinomas. Am. J. Surg. Pathol. 31(5), 653–663 (2007).

50.

51.

52.

53.

54.

55.

56.

57.



58.

59.

••

Wishart DS, Tzur D, Knox C et al.: HMDB: the Human Metabolome Database. Nucleic Acids Res. 35(Database issue), D521–D526 (2007). Griffin JL, Nicholls AW: Metabolomics as a functional genomic tool for understanding lipid dysfunction in diabetes, obesity and related disorders. Pharmacogenomics 7(7), 1095–1107 (2006). Lindon JC, Holmes E, Nicholson JK: Metabonomics in pharmaceutical R&D. FEBS J. 274(5), 1140–1151 (2007). Mol BW, Boll D, de Kanter M et al.: Distinguishing the benign and malignant adnexal mass: an external validation of prognostic models. Gynecol. Oncol. 80(2), 162–167 (2001). Farhi J, Jacobs HS: Early prediction of ovarian multifollicular response during ovulation induction in patients with polycystic ovary syndrome. Fertil. Steril. 67(3), 459–462 (1997). Gallion H, van Nagell JR Jr, Donaldson ES, Hanson MB, Powell DF: Immature teratoma of the ovary. Am. J. Obstet. Gynecol. 146(4), 361–365 (1983). Suzuki M, Tamura N, Kobayashi H et al.: Clinical significance of combined use of macrophage colony-stimulating factor and squamous cell carcinoma antigen as a selective diagnostic marker for squamous cell carcinoma arising in mature cystic teratoma of the ovary. Gynecol. Oncol. 77(3), 405–409 (2000). Einhorn N, Knapp RC, Bast RC, Zurawski VR Jr: CA 125 assay used in conjunction with CA 15–3 and TAG-72 assays for discrimination between malignant and non-malignant diseases of the ovary. Acta Oncol. 28(5), 655–657 (1989). One of the first studies emphasizing the significance of combined assays for ovarian cancer screening. Hayashi H, Yaginuma Y, Kitamura S et al.: Bilateral oophorectomy in asymptomatic women over 50 years old selected by ovarian cancer screening. Gynecol. Obstet. Invest. 47(1), 58–64 (1999). Buys SS, Partridge E, Greene MH et al.: Ovarian cancer screening in the Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening trial: findings from the initial screen of a randomized trial. Am. J. Obstet. Gynecol. 193(5), 1630–1639 (2005). One of the largest multicentered, randomized clinical trials evaluating ovarian cancer screening.

Women's Health (2008) 4(1)

60.



61.

62.

63.

64.

65.

66.

67.

68.

69.

70.

Menon U, Skates SJ, Lewis S et al.: Prospective study using the risk of ovarian cancer algorithm to screen for ovarian cancer. J. Clin. Oncol. 23(31), 7919–7926 (2005). Provides the first data regarding the use of risk of ovarian cancer algorithm in ovarian cancer screening. Skates SJ, Xu FJ, Yu YH et al.: Toward an optimal algorithm for ovarian cancer screening with longitudinal tumor markers. Cancer 76(Suppl. 10), 2004–2010 (1995). Boulman N, Levy Y, Leiba R et al.: Increased C-reactive protein levels in the polycystic ovary syndrome: a marker of cardiovascular disease. J. Clin. Endocrinol. Metab. 89(5), 2160–2165 (2004). Zhang Z, Bast RC, Jr., Yu Y et al.: Three biomarkers identified from serum proteomic analysis for the detection of early stage ovarian cancer. Cancer Res. 64(16), 5882–5890 (2004). Skates SJ, Horick N, Yu Y et al.: Preoperative sensitivity and specificity for early-stage ovarian cancer when combining cancer antigen CA-125II, CA 15–3, CA 72–4, and macrophage colony-stimulating factor using mixtures of multivariate normal distributions. J. Clin. Oncol. 22(20), 4059–4066 (2004). Huang CY, Cheng WF, Lee CN et al.: Serum mesothelin in epithelial ovarian carcinoma: a new screening marker and prognostic factor. Anticancer Res. 26(6C), 4721–4728 (2006). Badgwell D, Lu Z, Cole L et al.: Urinary mesothelin provides greater sensitivity for early stage ovarian cancer than serum mesothelin, urinary hCG free β subunit and urinary hCG β core fragment. Gynecol. Oncol. 106(3), 490–497 (2007). Wang P, Wu X, Chen W, Liu J, Wang X: The lysophosphatidic acid (LPA) receptors their expression and significance in epithelial ovarian neoplasms. Gynecol. Oncol. 104(3), 714–720 (2007). Sutphen R, Xu Y, Wilbanks GD et al.: Lysophospholipids are potential biomarkers of ovarian cancer. Cancer Epidemiol. Biomarkers Prev. 13(7), 1185–1191 (2004). Rosen DG, Wang L, Atkinson JN et al.: Potential markers that complement expression of CA125 in epithelial ovarian cancer. Gynecol. Oncol. 99(2), 267–277 (2005). Hellstrom I, Raycraft J, Hayden-Ledbetter M et al.: The HE4 (WFDC2) protein is a biomarker for ovarian carcinoma. Cancer Res. 63(13), 3695–3700 (2003).

future science group

Biomarker discovery in ovarian cancer – REVIEW

71.

72.

73.

• 74. 75.

76.

77.

78.

79.

80.

81.

82.

Bast RC Jr, Badgwell D, Lu Z et al.: New tumor markers: CA125 and beyond. Int. J. Gynecol. Cancer 15(Suppl.), 3274–3281 (2005). 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. 10(11), 3919–3926 (2004). 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. 18(8), 1733–1739 (2000). Important paper that describes the use of CA125 to define response in clinical trials. Schuijer M, Berns EM: TP53 and ovarian cancer. Hum. Mutat. 21(3), 285–291 (2003). 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. 63(6), 1311–1316 (2003). Safaei R: Role of copper transporters in the uptake and efflux of platinum containing drugs. Cancer Lett. 234(1), 34–39 (2006). Mansouri A, Zhang Q, Ridgway LD, Tian L, Claret FX: Cisplatin resistance in an ovarian carcinoma is associated with a defect in programmed cell death control through XIAP regulation. Oncol. Res. 13(6–10), 399–404 (2003). Hille S, Rein DT, Riffelmann M et al.: Anticancer drugs induce mdr1 gene expression in recurrent ovarian cancer. Anticancer Drugs. 17(9), 1041–1044 (2006). Wang Y, O’Brate A, Zhou W, Giannakakou P: Resistance to microtubule-stabilizing drugs involves two events: β-tubulin mutation in one allele followed by loss of the second allele. Cell Cycle 4(12), 1847–1853 (2005). Komatsu M, Hiyama K, Tanimoto K et al.: Prediction of individual response to platinum/paclitaxel combination using novel marker genes in ovarian cancers. Mol. Cancer Ther. 5(3), 767–775 (2006). Hunter VJ, Daly L, Helms M et al.: The prognostic significance of CA 125 half-life in patients with ovarian cancer who have received primary chemotherapy after surgical cytoreduction. Am. J. Obstet. Gynecol. 163(4 Pt 1), 1164–1167 (1990). Bonafe N, Gilmore-Hebert M, Folk NL et al.: Glyceraldehyde-3-phosphate dehydrogenase binds to the AU-Rich 3´ untranslated region of colony-stimulating factor-1 (CSF-1) messenger RNA in human ovarian cancer cells: possible role in CSF-1 posttranscriptional regulation and tumor phenotype. Cancer Res. 65(9), 3762–3771 (2005).

future science group

83.

84.

85.

86.

87.

88.

89.

90.

91.

92.

93.

Toy EP, Chambers JT, Kacinski BM, Flick MB, Chambers SK: The activated macrophage colony-stimulating factor (CSF-1) receptor as a predictor of poor outcome in advanced epithelial ovarian carcinoma. Gynecol. Oncol. 80(2), 194–200 (2001). Price FV, Chambers SK, Chambers JT et al.: Colony-stimulating factor-1 in primary ascites of ovarian cancer is a significant predictor of survival. Am. J. Obstet. Gynecol. 168(2), 520–527 (1993). Kleinberg L, Florenes VA, Silins I et al.: Nuclear expression of survivin is associated with improved survival in metastatic ovarian carcinoma. Cancer 109(2), 228–238 (2007). 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 98(1), 66–73 (2003). Sasaki N, Kudoh K, Kita T et al.: Effect of HER-2/neu overexpression on chemoresistance and prognosis in ovarian carcinoma. J. Obstet. Gynaecol. Res. 33(1), 17–23 (2007). Lenhard M, Kuemper C, Ditsch N et al.: Use of novel serum markers in clinical follow-up of Sertoli–Leydig cell tumours. Clin. Chem. Lab. Med. 45(5), 657–661 (2007). Patterson DM, Rustin GJ: Controversies in the management of germ cell tumours of the ovary. Curr. Opin. Oncol. 18(5), 500–506 (2006). Tsukishiro S, Suzumori N, Nishikawa H, Arakawa A, Suzumori K: Use of serum secretory leukocyte protease inhibitor levels in patients to improve specificity of ovarian cancer diagnosis. Gynecol. Oncol. 96(2), 516–519 (2005). Hogdall EV, Hogdall CK, Tingulstad S et al.: Predictive values of serum tumour markers tetranectin, OVX1, CASA and CA125 in patients with a pelvic mass. Int. J. Cancer 89(6), 519–523 (2000). van Haaften-Day C, Shen Y, Xu F et al.: OVX1, macrophage-colony stimulating factor, and CA-125-II as tumor markers for epithelial ovarian carcinoma: a critical appraisal. Cancer 92(11), 2837–2844 (2001). Moore LE, Fung ET, McGuire M et al.: Evaluation of apolipoprotein A1 and posttranslationally modified forms of transthyretin as biomarkers for ovarian cancer detection in an independent study population. Cancer Epidemiol. Biomarkers Prev. 15(9), 1641–1646 (2006).

www.futuremedicine.com

94.

95.

96.

97.

98.

99.

100.

101.

102.

103.

104.

Kozak KR, Su F, Whitelegge JP et al.: Characterization of serum biomarkers for detection of early stage ovarian cancer. Proteomics 5(17), 4589–4596 (2005). Simon I, Liu Y, Krall KL et al.: Evaluation of the novel serum markers B7-H4, Spondin 2, and DcR3 for diagnosis and early detection of ovarian cancer. Gynecol. Oncol. 106(1), 112–118 (2007). 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. 106(2), 334–341 (2007). Yesilada E, Sahin I, Ozcan H et al.: Increased micronucleus frequencies in peripheral blood lymphocytes in women with polycystic ovary syndrome. Eur. J. Endocrinol. 154(4), 563–568 (2006). Malamou-Mitsi V, Crikoni O, Timotheadou E et al.: Prognostic significance of HER-2, p53 and Bcl-2 in patients with epithelial ovarian cancer. Anticancer Res. 27(2), 1157–1165 (2007). Sorak M, Arsenijevic S, Lukic G et al.: Relationship of serum levels of tumor markers with tissue expression of gene products in ovarian carcinoma. J. Buon. 12(1), 99–104 (2007). Kikkawa F, Nawa A, Kajiyama H et al.: Clinical characteristics and prognosis of mucinous tumors of the ovary. Gynecol. Oncol. 103(1), 171–175 (2006). Gronlund B, Hogdall EV, Christensen IJ et al.: Pre-treatment prediction of chemoresistance in second-line chemotherapy of ovarian carcinoma: value of serological tumor marker determination (tetranectin, YKL-40, CASA, CA 125). Int. J. Biol. Markers. 21(3), 141–148 (2006). Sehouli J, Akdogan Z, Heinze T et al.: Preoperative determination of CASA (Cancer Associated Serum Antigen) and CA-125 for the discrimination between benign and malignant pelvic tumor mass: a prospective study. Anticancer Res. 23(2A), 1115–1118 (2003). 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. 10(10), 3291–3300 (2004). Heinzelmann-Schwarz VA, Gardiner-Garden M, Henshall SM et al.: Overexpression of the cell adhesion molecules DDR1, Claudin 3, and Ep-CAM in metaplastic ovarian epithelium and ovarian cancer. Clin. Cancer Res. 10(13), 4427–4436 (2004).

39

REVIEW – Tung, Wong & Mok

105. Samimi G, Safaei R, Katano K et al.:

106.

107.

108.

109.

110.

111.

112.

113.

40

Increased expression of the copper efflux transporter ATP7A mediates resistance to cisplatin, carboplatin, and oxaliplatin in ovarian cancer cells. Clin. Cancer Res. 10(14), 4661–4669 (2004). Samimi G, Varki NM, Wilczynski S et al.: Increase in expression of the copper transporter ATP7A during platinum drug-based treatment is associated with poor survival in ovarian cancer patients. Clin. Cancer Res. 9(16 Pt 1), 5853–5859 (2003). Lin YG, Han LY, Kamat AA et al.: EphA2 overexpression is associated with angiogenesis in ovarian cancer. Cancer 109(2), 332–340 (2007). Thaker PH, Deavers M, Celestino J et al.: EphA2 expression is associated with aggressive features in ovarian carcinoma. Clin. Cancer Res. 10(15), 5145–5150 (2004). Kang S, Ju W, Kim JW et al.: Association between excision repair crosscomplementation group 1 polymorphism and clinical outcome of platinum-based chemotherapy in patients with epithelial ovarian cancer. Exp. Mol. Med. 38(3), 320–324 (2006). 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. 25(16), 2281–2287 (2007). Gan Y, Wientjes MG, Au JL: Expression of basic fibroblast growth factor correlates with resistance to paclitaxel in human patient tumors. Pharm. Res. 23(6), 1324–1331 (2006). Osada R, Horiuchi A, Kikuchi N et al.: Expression of hypoxia-inducible factor 1α, hypoxia-inducible factor 2α, and von Hippel-Lindau protein in epithelial ovarian neoplasms and allelic loss of von HippelLindau gene: nuclear expression of hypoxia-inducible factor 1α is an independent prognostic factor in ovarian carcinoma. Hum. Pathol. 38(9), 1310–1320 (2007). Takao M, Okamoto A, Nikaido T et al.: Increased synthesis of indoleamine-2,3dioxygenase protein is positively associated with impaired survival in patients with serous-type, but not with other types of, ovarian cancer. Oncol. Rep. 17(6), 1333–1339 (2007).

114. Okamoto A, Nikaido T, Ochiai K et al.:

115.

116.

117.

118.

119.

120.

121.

122.

123.

124.

Indoleamine 2,3-dioxygenase serves as a marker of poor prognosis in gene expression profiles of serous ovarian cancer cells. Clin. Cancer Res. 11(16), 6030–6039 (2005). Maatta M, Talvensaari-Mattila A, Turpeenniemi-Hujanen T, Santala M: Matrix metalloproteinase-2 (MMP-2) and -9 (MMP-9) and their tissue inhibitors (TIMP-1 and TIMP-2) in differential diagnosis between low malignant potential (LMP) and malignant ovarian tumours. Anticancer Res. 27(4C), 2753–2758 (2007). Sillanpaa S, Anttila M, Voutilainen K et al.: Prognostic significance of matrix metalloproteinase-9 (MMP-9) in epithelial ovarian cancer. Gynecol. Oncol. 104(2), 296–303 (2007). Samanta AK, Huang HJ, Bast RC Jr, Liao WS: Overexpression of MEKK3 confers resistance to apoptosis through activation of NFκB. J. Biol. Chem. 279(9), 7576–7583 (2004). Wang L, Ma J, Liu F et al.: Expression of MUC1 in primary and metastatic human epithelial ovarian cancer and its therapeutic significance. Gynecol. Oncol. 105(3), 695–702 (2007). Nomura H, Tamada Y, Miyagi T et al.: Expression of NEU3 (plasma membrane-associated sialidase) in clear cell adenocarcinoma of the ovary: its relationship with T factor of pTNM classification. Oncol. Res. 16(6), 289–297 (2006). Ouellet V, Le Page C, Guyot MC et al.: SET complex in serous epithelial ovarian cancer. Int. J. Cancer 119(9), 2119–2126 (2006). Schorge JO, Drake RD, Lee H et al.: Osteopontin as an adjunct to CA125 in detecting recurrent ovarian cancer. Clin. Cancer Res. 10(10), 3474–3478 (2004). Nakae M, Iwamoto I, Fujino T et al.: Preoperative plasma osteopontin level as a biomarker complementary to carbohydrate antigen 125 in predicting ovarian cancer. J. Obstet. Gynaecol. Res. 32(3), 309–314 (2006). Baron AT, Boardman CH, Lafky JM et al.: Soluble epidermal growth factor receptor (sEGFR) [corrected] and cancer antigen 125 (CA125) as screening and diagnostic tests for epithelial ovarian cancer. Cancer Epidemiol. Biomarkers Prev. 14(2), 306–318 (2005). Secord AA, Darcy KM, Hutson A et al.: Co-expression of angiogenic markers and associations with prognosis in advanced epithelial ovarian cancer: a Gynecologic Oncology Group study. Gynecol. Oncol. 106(1), 221–232 (2007).

Women's Health (2008) 4(1)

125. Kuhn W, Marx D, Meidel A et al.:

126.

127.

128.

129.

130.

131.

132.

133.

134.

135.

Borderline tumors of the ovary: a clinicopathologic and immunohistochemical study of 54 cases. J. Obstet. Gynaecol. Res. 24(6), 437–445 (1998). Mills GB, Bast RC Jr, Srivastava S: Future for ovarian cancer screening: novel markers from emerging technologies of transcriptional profiling and proteomics. J. Natl Cancer Inst. 93(19), 1437–1439 (2001). Tsigkou A, Marrelli D, Reis FM et al.: Total inhibin is a potential serum marker for epithelial ovarian cancer. J. Clin. Endocrinol. Metab. 92(7), 2526–2531 (2007). Baba T, Mori S, Matsumura N et al.: Trophinin is a potent prognostic marker of ovarian cancer involved in platinum sensitivity. Biochem. Biophys. Res. Commun. 360(2), 363–369 (2007). Paju A, Vartiainen J, Haglund C et al.: Expression of trypsinogen-1, trypsinogen-2, and tumor-associated trypsin inhibitor in ovarian cancer: prognostic study on tissue and serum. Clin. Cancer Res. 10(14), 4761–4768 (2004). Mozzetti S, Ferlini C, Concolino P et al.: Class III β-tubulin overexpression is a prominent mechanism of paclitaxel resistance in ovarian cancer patients. Clin. Cancer Res. 11(1), 298–305 (2005). Ahmed AS, Dew T, Lawton FG et al.: M2-PK as a novel marker in ovarian cancer. A prospective cohort study. Eur. J. Gynaecol. Oncol. 28(2), 83–88 (2007). Orbo A, Hanevik M, Jaeger R, van Heusden S, Sager G: Urinary cyclic GMP after treatment of gynecological cancer. A prognostic marker of clinical outcome. Anticancer Res. 27(4C), 2591–2596 (2007). Muller-Klingspor V, Hefler L, Obermair A et al.: Prognostic value of β1-integrin (=CD29) in serous adenocarcinomas of the ovary. Anticancer Res. 21(3C), 2185–2188 (2001). Dupont J, Tanwar MK, Thaler HT et al.: Early detection and prognosis of ovarian cancer using serum YKL-40. J. Clin. Oncol. 22(16), 3330–3339 (2004). Dehn H, Hogdall EV, Johansen JS et al.: Plasma YKL-40, as a prognostic tumor marker in recurrent ovarian cancer. Acta Obstet. Gynecol. Scand. 82(3), 287–293 (2003).

Websites 201. American cancer society

www.cancer.org 202. UK Collaborative Trial of Ovarian Cancer

Screening www.ukctocs.org.uk/

future science group