Gynecologic Oncology 124 (2012) 354–365
Contents lists available at SciVerse ScienceDirect
Gynecologic Oncology journal homepage: www.elsevier.com/locate/ygyno
Review
Genetic polymorphisms as predictive and prognostic biomarkers in gynecological cancers: A systematic review☆,☆☆ Ivan Diaz-Padilla a, b,⁎, Eitan Amir a, Sharon Marsh c, Geoffrey Liu a, Helen Mackay a a b c
Division of Medical Oncology, Princess Margaret Hospital, University of Toronto, Ontario, Canada Centro Integral Oncologico Clara Campal, Madrid, Spain Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
a r t i c l e
i n f o
Article history: Received 6 October 2011 Accepted 29 October 2011 Available online 6 November 2011 Keywords: Gene polymorphism Ovarian cancer Predictive biomarker Gynecological cancer Vascular endothelial growth factor ERCC1
a b s t r a c t Purpose. Numerous studies have explored the potential role of genetic polymorphisms as predictive or prognostic biomarkers in gynecologic malignancies. A systematic review for all eligible polymorphisms has not yet been reported. The aim of this study was to summarize the current status of the field and provide direction for future research. Design. We searched literature databases (MEDLINE, EMBASE, Cochrane) from 2006 to April 2011 to identify studies evaluating the association between gene polymorphisms and clinical outcome in ovarian, endometrial, cervical, or vulvar cancer. The main outcome measures were overall survival (OS) and progressionfree survival (PFS). Studies reporting relationships between polymorphisms and toxicity were also included. Results. Sixty two studies met the inclusion criteria. The median sample size was 140. Most of the included studies (n = 50, 81%) were conducted in ovarian cancer patients. Almost a third assessed potential predictive associations between gene polymorphism and outcome in ovarian cancer. The most commonly evaluated genes were ERCC1, VEGF, ABCB1 (MDR), and GSTP1. Most studies (n = 44, 71%) were observational caseseries. Only four studies (6%) included a validation arm and patient population ethnicity was explicitly stated only in 27% of included studies. Conclusion. No consistent association between any gene polymorphism and clinical outcome in gynecological cancers has been found across studies. There is incomplete adherence to the REMARK guidelines and inadequate methodology reporting in most studies. Moving forward, analysis of large trial-based clinical samples; adherence to the highest methodological standards, and focus on validation analyses are necessary to identify clinically useful pharmacogenomic biomarkers of outcome. © 2011 Elsevier Inc. All rights reserved.
Contents Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Material and methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Search strategy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Study selection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Data extraction and management . . . . . . . . . . . . . . . . . . . . . . . Assessment of risk of bias in included studies . . . . . . . . . . . . . . . . . . Results . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Gene polymorphisms and ovarian cancer . . . . . . . . . . . . . . . . . . . . Non-VEGF pathway polymorphisms as predictive biomarkers of survival . . . . . Non-VEGF pathway polymorphisms as predictive biomarkers of toxicity in ovarian Non-VEGF pathway polymorphisms as prognostic biomarkers in ovarian cancer .
. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . cancer . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
. . . . . . . . . . .
355 355 355 355 355 355 356 356 357 357 357
☆ Support: I.D.P. is supported by the Programa de Formacion Avanzada en Oncologia at the Asociación Española Contra el Cáncer (AECC), Spain. G.L. is supported by the Alan Brown Chair in Molecular Genomics, the Poslun Family Foundation, Cancer Care Ontario (CCO) Chair in Experimental Therapeutics and Population Studies. ☆☆ Disclaimer: The authors declare no conflict of interest for the present manuscript. This study has not been presented in part or in whole elsewhere. ⁎ Corresponding author at: Division of Medical Oncology and Hematology, Princess Margaret Hospital, 610 University Avenue, Toronto, Ontario, Canada, M5G 2M9. Fax: + 1 416 946 2016. E-mail address:
[email protected] (I. Diaz-Padilla). 0090-8258/$ – see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.ygyno.2011.10.034
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Polymorphisms of the VEGF pathway and outcome in ovarian cancer . . . Polymorphisms and Outcome in Endometrial, Cervical, and Vulvar Cancers. Discussion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
Introduction
Material and methods
Gynecologic malignancies are a major cause of morbidity and mortality. Endometrial and ovarian cancer are the fourth and fifth most common malignancies diagnosed in North American women, while cervical cancer is the second most common worldwide [1]. Despite response rates of over 50% to frontline chemotherapy, relapse rates are high for all gynecologic cancers and recurrent disease incurable [2–4]. Optimizing the use of existing therapies, improving quality of life and the rational and cost-effective incorporation of emerging biological agents are all areas of high priority research. Identification of both prognostic and predictive biomarkers would be highly desirable to help achieve these goals. Validated prognostic biomarkers have the potential to improve risk stratification, thereby allowing more aggressive strategies for patients at risk of a poor outcome or, conversely, the adoption of less aggressive treatment for those whose risk of recurrence is lower. Identification of biomarkers which predict response (or the lack of it) to conventional or targeted treatment will allow patients to avoid toxicity from ineffective therapy and to be fast-tracked into clinical trials. Furthermore, the identification of biomarkers which predict particular drug-related toxicities, for example hypertension associated with anti-angiogenic agents, [5] would allow pro-active preventative measures to be taken in those at risk. Germline polymorphisms are heritable variations in the human genome that typically occur in 1% or greater frequency in the population being studied. Although much of the literature has focused on the association between genetic polymorphisms and the susceptibility of disease (i.e., case–control studies comparing cancer patients to healthy normal individuals), more recently the focus has shifted to include evaluations of the relationships between these variants and clinical outcome after cancer diagnosis [6]. These outcomes include survival parameters (e.g., overall survival [OS], progression or disease free survival [PFS/DFS] and time-to-progression [TTP]), response rates (RR), and acute or chronic toxicity of therapies. Two distinct roles of polymorphism relationships are evaluable: (i) genetic variants as predictive markers of treatment outcome, whereby these biomarkers predict one set of outcomes in patients receiving specific therapies, but have no relationship (or perhaps a completely different relationship) in the absence of receiving such therapies (pharmacogenomics) and (ii) polymorphisms as prognostic markers of outcome, whereby the relationship between the biomarker and clinical outcome is independent of any treatment aspects. In August 2010 the Gynecologic Cancer Intergroup (GCIG) recognized the importance of exploring and promoting this approach and formed the GCIG Ovarian Cancer Pharmacogenomics Working Group. Although numerous pharmacogenomic studies have been reported in gynecologic cancer, most commonly in women with ovarian cancer, to date there has been no systematic review of the published literature to guide future research. Furthermore, as platinum and taxane-based chemotherapy combinations are standard of care for the major gynecologic malignancies, [7–9] any potentially predictive biomarker signature which emerges in one population of women may be relevant for future studies in other gynecologic tumor populations. The goals of this review are three-fold: (i) to summarize the state of the current research field; (ii) to identify the top candidate choices that are either validated, or worthy of ongoing validation; and (iii) to identify gaps and direct the future research in this field.
Search strategy
355
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
. . . . . .
363 363 363 363 364 364
Literature searches of MEDLINE (host: OVID), EMBASE (host: OVID) and the Cochrane Central Register of Controlled Trials, from 2006 to April 2011; were performed using keywords and MeSH terms (Appendix 1). The search was restricted to English language articles reporting human studies. Citation and reference lists of retrieved articles were checked to ensure sensitivity of the search strategy. Authors of relevant studies were contacted and asked if they knew of further data which may or may not have been published. Attempts were made to contact and clarify ambiguous result reporting from corresponding authors, where necessary. Study selection Titles and abstracts were retrieved and assessed. Commentaries, single case-reports, editorials, review articles, unrelated articles and duplicates were excluded (authors I.D.P. and G.L.). Full-text articles were obtained for the remainder. The eligibility of retrieved papers was assessed independently by three review authors (I.D.P, G.L., H. M.). English language articles published in peer-reviewed journals that assessed the relationship between germline polymorphic variants and major outcomes of interest (hazard ratio [HR] for survival, odds ratio [OR] for toxicity) were included (Fig. 1). Data extraction and management For included studies, the following data were abstracted: author, year of publication (if published), journal citation, country, inclusion and exclusion criteria, study design and methodology; distribution of clinicopathologic factors (including age, histology, tumor grade, International Federation of Gynecology and Obstetrics [FIGO] stage, extent of residual disease), intervention details (type, timing and outcome of surgery, chemotherapeutic agents, and radiation therapy for non-ovarian studies); duration and completeness of follow-up; genotyping material, methods and completeness; analytical strategy (including whether multivariate analyses were performed) and associations between polymorphisms and outcome data. Relationships with surrogate and unconventional outcomes were excluded. Assessment of risk of bias in included studies The risk of bias in the included randomized clinical trials was assessed using the Cochrane Collaboration's tool and criteria [10]. The risk of bias in non-randomized studies was assessed in accordance with additional criteria: (i) study design; (ii) criteria for assignment of patients to treatment (where provided); (iii) whether or not patients included in analyses were representative of the larger population who would be treated for this condition (external validity); and iv) whether or not bias within the study design and analysis (i.e. internal validity) were appropriately considered. For example, in ovarian cancer, the key prognostic factors such as International Federation of Gynecology and Obstetrics (FIGO) stage, histology, type and extent of surgery, and type of treatment were chosen as minimally acceptable factors required to be considered in analyses.
356
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Literature Search Databases: Pubmed (2006-April 2011) EMBASE (2006-April 2011) Cochrane Library (2006-April 2011) English -language articles only
Search results combined (n=1462)
292 Duplicates
1170 Manuscripts screened on basis of title and abstract 1108 Manuscripts excluded 780 Articles of unrelated topic 328 Risk/susceptibility studies
62 manuscripts included Fig. 1. Flow diagram of the literature search and selection of included studies.
Three authors (I.D.P, G.L., H.M.) reviewed all papers independently; differences were resolved by consensus. Results A summary of included studies is shown in Table 1. The literature search found 1462 articles. After application of eligibility criteria, 62 studies were identified as evaluating the association between gene
Table 1 Characteristics of included studies. Study characteristics
No. of studies (%)
Study design Case series Case–control Randomized phase III clinical trial Population registry Phase II clinical trial
44 (71) 11 (18) 4 (7) 2 (3) 1 (2)
Studies by tumor type Ovarian Cervical Endometrial Vulvar
50 (81) 8 (13) 3 (5) 1 (2)
Biospecimen source for genotyping Blood leukocytes FFPE tumor Fresh tumor tissue Not specified
40 (64) 14 (23) 5 (8) 3 (5)
Statistical analysis Multivariate Univariate Total no. of included studies
46 (74) 16 (26) 62 (100)
Abbreviations: FFPE, formalin-fixed paraffin-embedded.
polymorphisms and clinical outcome in ovarian, endometrial, cervical, or vulvar cancer (Fig. 1). The majority of these studies were either case series (n = 44, 72%) or cases that formed part of an existing case– control study (n = 11, 18%). Other study designs included cohort observational studies or subset analyses of existing cohort studies (n = 2, 3%), or secondary analyses of randomized clinical trials (n = 4, 7%). Only four studies (6%) included a validation group. Sixteen studies (26%) did not report multivariate analysis in their results. While the majority of studies (n = 40, 64%) performed the genotyping analysis on blood-derived specimens, fourteen studies (23%) performed genotyping on formalin-fixed paraffin-embedded material, five (8%) used frozen tumor tissue, and in three studies (5%) the source for genotyping was not clearly stated. Study populations were predominantly Caucasian (N = 50, 81%), reported or inferred based on the academic affiliation of authors or hospital location. For those studies that explicitly stated the ethnicity of the patient population (n = 17, 27%), no uniform categorization for the non-caucasian cohort (i.e. African American, Hispanic) was found across studies. Twenty five studies (40%) provided little to no information on the treatment their patients received. Though we attempted to contact corresponding authors about missing information, in many cases, no additional information was obtained. For study size assessment, we reported only on the subset of individuals who had genotyping attempted. Study size ranged from 16 to 1499 patients, with a median of 140 (mean of 247). Almost all studies identified themselves as exploratory or in need of validation or replication.
Gene polymorphisms and ovarian cancer Fifty studies evaluated the association between polymorphisms and outcome in ovarian cancer. We separated studies into those that were predictive (Table 2) or prognostic (Table 3 and Supplementary Table 1). Because of the large number of prognostic and
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
predictive studies evaluating the vascular endothelial growth factor (VEGF) pathway, we collated all such studies, regardless of whether they had significant associations or not (Table 4). Most studies reported polymorphisms in multiple genes, as well as different polymorphisms within the same gene. For those studies where the same polymorphism was evaluated, the outcome measures were different and/or the hazard ratios were not available, making it difficult to perform a pooled analysis data or to assess for heterogeneity. Non-VEGF pathway polymorphisms as predictive biomarkers of survival A summary of included studies [11–29] is shown in Table 2. Six studies of adjuvant platinum and taxane-based therapy assessed the association between polymorphisms in the P-glycoprotein (PGP) gene, ABCB1 (MDR1), and treatment outcome [13, 14, 16, 19, 22, 25]. Taxanes and other cytotoxic drugs can be extruded from the cell by P-glycoprotein (PGP), an ATP-driven drug export pump. Its overexpression is associated with drug-resistant phenotypes [30]. Results from ovarian cancer clinical studies suggest that increased PGP expression, partly influenced by the ABCB1 polymorphisms 2677G>T/A and 3435C>T, [31] is associated with poorer outcome [32]. Of the three studies that included multivariate analyses, [16, 19, 22] only one study found borderline significant associations with 2677G>T/A, [16] while the largest analysis that used samples from a randomized phase III trial (SCOTROC1) found no association [22]. None reported associations between ABCB1 3435C>T and outcome. Nine studies evaluated the predictive nature of polymorphisms in the nucleotide excision DNA repair pathway with platinum therapy [17–22, 26–28]. Eight studies reported two common polymorphisms of the excision repair cross-complementation group 1 (ERCC1) gene, which codes the protein that repairs cisplatin-induced DNA damage. The ERCC1 codon 118 polymorphism features a C to T transition, but does not change the amino acid, asparagine; the T variant however has been associated with a 50% reduction in transcription, and in an ovarian cancer cell line, has been linked to decreased levels of ERCC1 mRNA, indicating reduced DNA repair capability [33]. In this review, data for the ERCC1 codon 118 polymorphism has been conflicting. Three studies (two case-series studies [17, 27] and one phase III clinical trial [21]) reported the C/C genotype to be associated with higher risk of platinum resistance, disease progression, and death from ovarian cancer; while two analyses of other prospective clinical trials did not confirm such results [20, 22]. The A allele of the second polymorphism, ERCC1 8092C>A, located in the 3′ untranslated region, may affect mRNA stability, and has been associated with more favorable outcomes in other solid tumors [34, 35]. However, ERCC1 8092 A/-genotypes were associated with poorer PFS and OS in two different phase III trials of platinum-based chemotherapy for optimally resected stage III ovarian cancer patients, [20, 21] and in a smaller retrospective study [19]. Other polymorphisms in the nucleotide excision repair pathway (ERCC2 (XPD), XPA, XPC, and XPG), and the base excision pathway (XRCC1) have also been evaluated, with varying associations reported (Table 2) [26]. The glutathione-S-transferase (GST) enzymes catalyze the conjugation of glutathione to platinum, [36] limiting the amount of free platinum drug available for interaction with DNA. Three GST genes, GSTP1, GSTM1 and GSTT1, have common polymorphisms associated with decreased enzymatic activity [37]. GSTP1 105 A>G was the only polymorphism evaluated across multiple studies, with conflicting results. In two studies, the variant G allele was associated with improved outcome, either alone or in combination with another GST polymorphism [11, 24]. This was not confirmed in other studies, [18, 23] while the largest analysis reported no association [22]. Other polymorphisms within genes involved in xenobiotic metabolism have been analyzed in two small studies [15, 29]. Only a polymorphism in the cytochrome P450 complex, namely CYP1A1 Ile462Val, has been associated with platinum resistance [15].
357
Non-VEGF pathway polymorphisms as predictive biomarkers of toxicity in ovarian cancer Four studies evaluated the association of non-VEGF pathway polymorphisms with toxicity in ovarian cancer [18, 19, 22, 29]. Three studies assessed the occurrence of grades 3 and 4 toxicities based on pre-specified criteria [19, 22, 29]. One study found borderline associations between polymorphisms (CDKN1A 10971C>T and CYP1B1*3) and increased risk of developing gastrointestinal toxicity when patients were treated with platinum-docetaxel in a training set. This association, however, was not confirmed in an internal validation set [22]. Two univariate analyses of smaller retrospective studies reported associations between polymorphisms in the DNA repair, multi-drug resistance, and drug metabolism pathways and a wide spectrum of toxicities [18, 19]. Kim et al. reported an increased risk of gastrointestinal toxicity for carriers of ABCB1 2677 variants, and a higher risk of hematological toxicity for those patients with a homozygous GSTP1 105 A/A genotype [19]. Khrunin et al. reported that patients with a homozygous GSTM1 deletion (GSTM1 null) experienced fewer adverse events (myelosuppresion, neuropathy) than those who carried functional GSTM1 variants [18]. None of the polymorphism-toxicity relationships has been validated in independent studies. Non-VEGF pathway polymorphisms as prognostic biomarkers in ovarian cancer Twenty five studies evaluated the potential prognostic value of non-VEGF polymorphisms in ovarian cancer. Of these, seven reported no significant associations [38–44] (Supplementary Table 1). Of the remaining studies with at least one positive association [45–62] (Table 3), the significant polymorphisms were from genes involved in a multitude of biological pathways, including tumor suppressor genes, oncogenes, microRNA (miRNA), inflammation, apoptosis, cellular senescence, detoxification, vitamin D, and xenobiotic metabolism. All published studies were hypothesis-generating. Two pathways reported significant associations across at least two studies: p53/cell cycle and hormone (estrogen and/or androgen) pathways. An intact p53 pathway is an important determinant of the response to platinum-based chemotherapy [63]. Polymorphisms and somatic genetic alterations of genes encoding p53 and its regulators have been shown to affect ovarian cancer pathogenesis [64]. Both the MDM2 and the MDM4 genes are critical negative regulators of p53 protein [65, 66]. It has been shown that the G-allele of the promoter MDM2 309T>G gene is associated with the attenuation of the p53 tumor suppressor pathway [65]. MDM2 expression is also regulated by the estrogen receptor (ER) pathway [67]. Three studies evaluated the p53-MDM pathway and found significant associations between clinical outcome and at least one MDM2, MDM4, or p53 polymorphism; in one case, the relationship was limited to ER-negative tumors [45, 58, 62]. Alterations in the androgen hormone homeostasis have been shown to influence the pathogenesis and progression of ovarian cancer either directly or indirectly [68, 69]. The androgen receptor (AR) is frequently overexpressed in ovarian cancer [70]. The AR gene contains a polymorphic cytosine–adenine–guanine (CAG) repeat in exon 1; the length of the CAG repeat sequence inversely correlates with AR function [71]. Two small studies examined the prognostic role of AR gene polymorphisms in ovarian cancer, and both found that the shorter AR alleles (each using different cut-offs) were associated with worse outcome [52, 54]. In contrast, this association was not replicated in a much larger study [55]. Nagle et al. found a significant association between a polymorphism of CYP17C, which codes for an enzyme involved with progesterone metabolism (CYP17 rs743572; 5′UTR T>C), but this association has not been externally validated.
358
Table 2 Ovarian cancer: gene polymorphisms with a predictive association with outcome.
MDR1
Country
N
Johnatty SEa (Population Registry) [16]
Australia
309 Carboplatin Paclitaxel
ABCB1 ABCB 1
1236C>T 2677G>T/A
Green Ha [13] (case series) Sweden
53
Carboplatin Paclitaxel
ABCB 1 ABCB 1
3435C>T 2677G>T/A
Green Ha [14] (case series) Sweden
51
Carboplatin Paclitaxel
Obata H [25] (case series)
Japan
44
Platinum-Based
ABCB 1 ABCB 1 ABCB 1 ABCB 1 MRP1 MRP2 ABCB 1
Marsh Sc [22] (phase III clinical trial)
United Kingdom
914 Platinum Taxane
ABCB 1 ABCB 1 ERCC1 ERCC2 XRCC1
3435C>T 1236C>T 1199G>T/A 1308A>G 2168G>A; 5 more SNPs 3 SNPs 1236C>T, 3435C>T, exon 28 A>G; 2 more SNPs 1236C>T 2677G>T/A, 3435C>T ^118C>T, 8092C>A 751 A>C 399G>A
GSTP1 TP53 ERCC1
Krivak TC [21] (phase III clinical trial)
ERCC1
USA
Treatment
280 Platinum Taxane
Gene(s) Polymorphism(s)
Subset analysis for ABCB 1 2677 and residual disease ≤1 cm: any variant vs GG: HR 0.6 (0.4–0.9) p = 0.01 Analysis was performed on frozen tumor tissue
All OS comparisons; p = NS ABCB1 2677T/T or T/A had greater chance of good response: *RR 1.8 (1.2–2.8); p = 0.04b ABCB1 1199 Carrying at least one A allele was associated with 2 vs 19 months OSb MRP1 2168 A/- vs G/G A/- had greater RR, p = 0.01 All comparisons; p = NS
No multivariate analysis No multivariate analysis No multivariate analysis
ERCC ^118T/- vs CC HR 0.7 (0.5–1.0); p = 0.03
Age FIGO stage Histology Residual disease
ERCC1
8092C>A
OS
NS
RR, OS
NS
PFS
ERCC1 8092 A/- vs C/C HR 1.9 (1.1–3.5); p = 0.03
RR
GSTT1 non-null vs -null, OR 0.3 (0.2–0.7); p = 0.04
OS
HR 1.7 (1.8–3.4)d; p = 0.04
Kim HS [19] (case series)
Korea
118 Platinum Taxane
ERCC1 GSTT1 ERCC2 GSTM1 GSTP1
8092C>A, ^118C>T -null 751 A>C -null 105A>G
ABCB1 XRCC1 ERCC1 XRCC1 XRCC1 GSTP1 GSTA1 ERCC2 TP53 ERCC1
2677G>T/A, 3435C>T R399Q, R194W ^118C>T, 8092C>A 399G>A, 280G>A 194C>T 105A>G, 114C>T -69C>T 312G>A,751 A>C Arg72Pro ^118C>T
103 Platinum-based
FIGO Stage Residual disease (1 cm)
OS
^118C>T
Italy
RR PFS
ABCB1 2677: others vs G/G HR 0.7 (0.5–1.0); p = 0.07
105 A>G, A114V Arg72Pro ^118C>T
ERCC1
Smith S [27] (case series)
PFS (no statistics reported)
Additional comments
Age Tumor grade FIGO stage Histology Residual disease (2 cm) ECOG PS
159 CarboplatinCyclophosphamide
104 Cisplatin Cyclophos-phamide
OS DFI>1 year vs DFIb1 year
Covariates considered
NS
Denmark
Russia
PFS
Reported association
RR, PFS
Steffensen KD [28] (phase III clinical trial)
Khrunin AVe [18] (case series)
Outcome measure
PFS
OS
PFS OS
GSTP1 105A>G A/A had better PFS; p = 0.0001 GSTA1-69C>T T/T had better OS; p = 0.05 ERCC2 312G>A, XPD 751 A>C: Hetero genotypes had Better PFS; p b 0.05 ERCC1 ^118C/C vs T/HR 2.0 (1.1–3.8) p = 0.05 HR 2.0 (1.1–3.8); p = 0.03
Tumor grade FIGO stage Histology Residual disease (1 cm) ERCC1 expression Age Tumor grade FIGO stage Histology Residual disease (1 cm)
Results based on only 2 of 51 patients carrying MDR 1199 A alleles Analysis was performed on formalin-fixed paraffin embedded (FFPE) specimens
ERCC 8092 A/- vs CC: trend toward increased risk of death [HR 1.3 (1.0–1.7); p = 0.08) Analysis was performed on FFPE specimens
No multivariate analysis
No association between genotype and response rates
Age Tumor grade FIGO stage
Combination of C/C+ high ERCC1 mRNA expression HR 3.7 (1.6–8.9); p = 0.03
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
MDR1 and ERCC1
Author (study type)
Table 2 (continued)
ERCC1, IP
ERCC2
Author (study type)
Country
N
Treatment
Gene(s) Polymorphism(s)
Kang S [17] (case series)
Korea
60
Carboplatin Paclitaxel
ERCC1
Krivak TC [20] (phase III clinical trial)
USA
Saldivar JS [26] (case series)
USA
Nagle CMg [24] (cases Xenobiotic metabolism from case–control) and p53
Italy
^118C>T Platinum resistance
233 Intraperitoneal (IP) vs Intravenous (IV) Cisplatin, Paclitaxel
ERCC1 ERCC1
^118C>T 8092C>A
125 Carboplatin Paclitaxel
XPA XPG XPC ERCC2 GSTP1
228A>G 1104A>H poly-AT 312G>A; 751A>C GSTP1 105A>G
454 Platinum-based (N = 340, 75%)
215 Platinum-based (N = 101, 47%)
Morari EC [23] (case series)
Brazil
69
Heubner M [15] (case series)
Germany
111 No data reported by authors
Gaducci A [12] (case series)
Italy
46
Takano M [29] (case series)
Japan
24
Platinum-based (N = 29, 42%)
Adjuvant Carboplatin Paclitaxel CisplatinIrinotecanh
All OS comparisons; p = NS
PFS OS
ERCC1 8092 A/- vs C/C, HR 1.4 (1.1–1.9), p = 0.02 HR 1.5 (1.1–2.1), p = 0.02
PFS OS
GSTT1 null GSTM1null GSTM1 null PFS OS
GSTP1 GSTT1
GSTP1 105A>G GSTT1 null
GSTP1 GSTT1 GSTM1 GSTO2 TP53 CYP1A1
GSTP1 105A>G GSTT1 null GSTM1null GSTO2 asn142asp Arg72Pro CYP1A1 Ile462Val
ERCC1 ^118T/- vs C/C OR 0.2 (0.0–0.7); p = 0.02
OS
OS GSTT1 GSTM1 GSTM1
Reported association
PFS OS, RR RR
Platinumresistance
XPA 228G/- vs A/A HR 9.1 (1.1–74); p = NR XPG 1104C/- vs G/G HR 8.9 (1.3–64); p = NR GSTP1 105G/- vs AA: HR 0.8 (0.6–1.0); p = 0.04
GSTM1 null vs non-null HR 0.7, CI 0.4–1.0, p = 0.02 HR 0.7, CI 0.5–1.0, p = 0.06 GSTM1 null/GSTP1 105G/- vs other HR 0.4, CI 0.2–0.8, p = 0.003 NS NS
CYP1A1 462 Val/- is associated with higher rate of platinum resistance OR 5.9 (1.5–23.2), p = 0.005
TP53
Arg72Pro
OS PFS
NS
UGT1A1
UGT1A1*28 UGT1A1*27 UGT1A1*6
RR
NS
Covariates considered
Additional comments
Histology Age Tumor grade
(PFS) Platinum-free interval defined as the interval between treatment commencement and date of relapse
FIGO stage Histology Residual disease (2 cm) Age Tumor grade FIGO stage Histology Age FIGO stage Histology
Residual diseasef GOG PS Ethnicity Treatment arm No significant associations with RR
Age Tumor grade FIGO stage Histology
Carriage of three low function GST genotypesc had trend toward better OS: HR 0.5 (0.2–1.0)
Age Tumor grade FIGO Stage Histology
Analysis was performed on frozen tumor tissue
Not reported by authors
Tumor grade FIGO stage
Analyses were performed on FFPE specimens
Residual disease Platinum resistance No multivariate analysis
Analyses were performed on FFPE specimens
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Beeghly A [11] (case series)
Australia
Outcome measure
No multivariate analysis
359
Studies in each subcategory were listed based on sample size (largest listed first). The first six studies listed evaluated ABCB1 polymorphisms. The fifth through thirteenth studies evaluated DNA Repair Polymorphisms, mainly on ERCC1. The Krivak et al. study was listed at the end because it involved intraperitoneal administration of chemotherapy, while Saldivar et al. did not include ERCC1. The five last studies focused on xenobiotic metabolism and p53 polymorphisms. The Morari et al. study was grouped within the studies evaluating GST polymorphisms. PFS, progression-free survival; OS, overall survival; DFI, disease-free interval; RR, response rate; *RR, Relative Risk; HR, hazard ratio; OR, odds ratio; NS, not significant; SNP, Single Nucleotide Polymorphism; FIGO, International Federation of Obstetrics and Gynecology; ECOG, Eastern Cooperative Oncology Group; PS Performance Status; GOG, Gynecologic Oncology Group; NR, not reported; IP, intraperitoneal. a Baseline population is virtually identical between these two studies. b Not adjusted. c Other polymorphisms evaluated in Marsh et al.: ABCC1 S1334S, ABCC1 IVS19-30C>G, ABCC2 24C>T, ABCC2 IVS12 + 148A>G, ABCC2 V417I, ABCG2 Q141K, CYP2C8 M264I, CYP2C8 R139K, CYP2C8 K399R, CYP3A4*1B, CYP3A4*3, CYP3A5*3C, ERCC1 17677G>T, MAPT P587P, MPO-463G>A, CDKN1A 10971C>T, CYP1B1*3. d The HR is outside of the 95% CI, authors were not able to resolve these contradictory numbers. e Other polymorphisms evaluated in Khrunin et al.: GSTM1 (gene deletion), GSTM3 (intron 6, gene deletion), GSTT1 (gene deletion), TP53 (intron 3, 16-bp dupl), TP53 (intron 6, rs1625895), CYP2E1rs2031920, CYP2E1rs6413432, CYP2E1rs2070676, CYP2E1 (5′flanking, 96-bp insertion). f Gross vs microscopic disease. g Also partially reported in Table 3. Baseline populations are identical between this study and that in Table 3. h Patients had recurrent disease. Also partially reported in Supplementary Table 2. ^ERCC1 codon 118.
360
Table 3 Ovarian cancer: gene polymorphisms (non-VEGF) with at least one positive prognostic association. Country
N
Treatment
Gene(s)
Polymorphism(s)
Outcome measure
Reported positive associations
Covariates considered
Additional comments
BRAF KRAS ERBB2 NMI PI3KCA CYP17 + othersd
BRAF rs6944385 and other 33 TagSNPs were genotypedb
OS
BRAF rs6944385 HR 1.2 (1.0–1.5); p = 0.01
Age Tumor grade FIGO stage Histology
BRAF Haplotype: HR 1.4, CI 1.15–1.8; p = 0.001; Associations not adjusted for multiple testing
CYP17 5′UTR T>C And other 20 SNPs 226 SNPs analyzed
OS
CYP17 5′UTR C/- vs TT HR 1.3 (1.0–1.7); p = 0.04
OS
PDGFC rs1425486 HR 2.7 (1.7–4.3); q = 0.004
Age FIGO stage Tumor grade Histology Age Histology FIGO stage Chemotherapy -adjusted for multiple comparisons Age Ethnicity Tumor grade Chemotherapy FIGO stage CA-125 Histology Place of surgery Residual disease
UK, USA, Quaye L [57] (cases from Denmark case–control) incident, prevalent cases combined
1480 No details
Nagle CMc [55] (cases from case–control) Liang D [53] (cases from case–control)
Australia
454
No details
USA
339
Platinum-based chemotherapy
USA
325
Platinum-taxane
142 miRNA pathway-related genes Multiple pathwaysf 1416 SNPs analyzed
Goode EL [48] (case series)
OS OS OS OS OS OS OS OS
Batra Jg [46] (cases from Australia case–control) Paige AJW [56] (case series) UK
319
No details
KLK15
235
No details
Hefler LA [50] (case series)
Austria
199
No details
WWOX WWOX, WWOX WWOX IL-6
Garg R [47] (case series)
USA
160
No details
IL-6
rs266851 and other 14 TagSNPs were genotypedb 1497T>G, 121C>T, Isnp1 C>T, PFS Isnp8 C>T, Isnp13 A>C. 660 PFS A>G, 1442C>T, Ispn15 A>G OS IL-6 174G>C PFS OS IL-6 174G>C OS
Han CH [49] (case series)
USA
136
Platinum-Taxane
Wynendaele J [62] (case series)
Germany
154
Platinum-based chemotherapy
SULF1 SULF1 SULF1 MDM4
rs2623047 G>A rs13264163, rs6990375, rs3802278, rs3087714 34091 A>C
Six L [60] (case series)
Austria
151
Platinum-based chemotherapy
PFS OS
OS PFS MMP1
− 1607 (G)/GG PFS OS
And for another 23 SNPse VHL rs265318 HR 0.7 (0.4–1); p = 0.04 EIF2B5 rs4912474 HR 0.7 (0.5–0.9); p = 0.004 CCR3 rs4987053 HR 1.6 (1.1–2.4); p = 0.01 IL1B rs1143634 HR 1.4 (1.0–1.8); p = 0.02 IL18 rs5744247 HR 0.5 (0.3–0.8); p = 0.003 IL18 rs11214108 HR 0.5 (0.3–0.9); p = 0.01 CYP1A1 rs2470893 HR 0.7 (CI 0.5–0.9); p = 0.005 KLK15 rs266851 T/- vs CC HR 1.4 (1.0–2.0), p = 0.05 WWOX 1497 GG vs TT: HR 2.1 (1.3–3.4); p = 0.003 GT vs TT: HR 1.6 (1.1–2.3); p = 0.01 P = NS IL-6 174C/- vs G/G was associated with improved PFS (p = 0.003) and OS (p = 0.02) IL-6 174G/G vs others was associated with 131 vs 28 months median OS, p = 0.001 SULF1 rs2623047 G/- was associated with improved PFS (28 vs 12 months, p = 0.02) Subgroup analysis of ER negative tumors MDM4 34091 A/A vs C/HR 5.5 (1.5–20); p = 0.01 HR 4.1 (1.2–14); p = 0.02 MMP1 others vs GG/GG HR 2.1, CI 1.2–3.8; p = 0.01 HR 1.9, CI 1.1–3.4; p = 0.04
Age FIGO stage Tumor grade Histology Age Association not confirmed Tumor grade in SCOTROC1, n = 863 FIGO stage Histology FIGO stage residual disease Lymph node metastases Age Histology Tumor grade Residual disease FIGO stage No multivariate analysis
Residual disease
Tumor grade FIGO stage
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Author (study type) populationa
Santos AM [58] (case series) Portugal
119
Platinum-based chemotherapy
Bartel F [45] (case series)
103
Platinum-based chemotherapy
Germany
TP53
Arg72Pro
PFS
P21 TP53 MDM2
3′UTR C>T Arg72Pro 309T>G
OS OS OS OS
Japan
101
No details
VDR
VDR FokI C>T
OS
Li AJ [52] (case series)
USA
77
No details
AR
CAG repeat length
OS
69
Platinum-based chemotherapy
AR AR FSH FSH PGRG CXCR2 AMg IL8 And morea
CAG repeat length GGN repeat length Ala307Thr Ser680Asn 331G>A 785C>T CA repeat -251T>A
LTBP-1L
202G>C 20 A>C
Ludwig AH [54] (cases from Poland case Control)
Schultheis AM [59] (single arm phase II clinical trial)
USA and Canada
53
CyclophosphamideBevacizumab
Higashi [51] (case series)
Japan
42
No details
PFS PFS OS
RR PFS PFS RR
OS
CXCR2 785T/T vs C/RR 2.1, CI 1.01–4.3; p = 0.03 AML≥14 repeats; S b 14 L/L vs S/S: HR 0.3; p = 0.04 L/S vs S/S: HR 0.5; p = 0.04 IL-8-251T>A: increasing number of A alleles was associated with lower RR (p = 0.006) Haplotype analysis: others vs G/A had longer 5-y OS (p = 0.02)
No multivariate analysis
FIGO stage Residual disease
Analysis were performed on FFPE specimens and not adjusted for multiple comparisons
Age FIGO stage Age Tumor grade FIGO Stage Age Tumor grade FIGO stage Histology
Histology Residual disease Analysis were performed on frozen tumor tissue Residual disease TP53 status Response to CT Platinum sensitivity
No adjustment for multiple comparisons
No multivariate analysis
Analysis were performed on FFPE specimens
Studies were listed from largest sample size to smallest. OS, overall survival; HR, Hazard Ratio; PFS, progression-free survival; q; false discovery rate adjusted p value; SNP, single nucleotide polymorphism; FIGO, International Federation of Gynecology and Obstetrics; NS, non-significant; CA-125, cancer antigen 125; FFPE, formalin-fixed paraffin embedded; CT, chemotherapy; miRNA, microRNA: small, non-coding RNA molecules involved in gene expression regulation; PDGFC, platelet-derived growth factor C; WWOX, WW-domain containing oxidoreductase; ER, estrogen receptor; MMP-1, Matrix metalloproteinase-1; VDR, vitamin D receptor; BMI, Body Mass Index; AR, androgen receptor; FSHR, follicule stimulating hormone receptor; PGRG, progesterone receptor gene; LTBP-1L, latent TGFβ- binding protein. a Other polymorphisms evaluated in Schultheis et al.: NRP1 3′UTR C>T; PGF (placental growth factor) 3′UTR rs8185; KDR 3′UTR T>A (rs12411) and AC repeat + 4422; LEP (Leptin) -2548G>A; CXCR1 2607G>C; CXCR2 785C>T; IGF2 4205G>A; IGFR exon 16 3174G>A; FGFR4 Gly288Arg; F3 (tissue factor) 5′UTR -603A>G; MMP2 -1306C>T; MMP9 -1562C>T; MMP7 -181 A>G; ICAM1 241G>A; EGF 61A>G; EGFR 497G>A; HIF1A 1772C>T; ARNT (HIF1B) rs2228099 G>C; IL6 174G>C; NFKB CA dinucleotide repeat; TNF -308G>A; IL1B -511T>C and 3954C>T; IL1R1 (IL-1Ra) intron 2, 86 bp repeat; MDM2 309T>G; P53 Arg72Pro; COX2 8473T>C; CXCL12 (SDF1) -801G>A. b TagSNPs utilize linkage disequilibrium to choose relatively independently sorting polymorphisms to represent as markers the genetic heritability of each segment of DNA across the chosen gen. c Baseline population is virtually identical between this and a corresponding study reported in Table 2. d Androgen receptor (AR) exon 1 CAG repeat (cutpoint ≥22) was also assessed, but no significant association was found. e The other 23 SNPs did not remain significant after adjusting for multiple testing. f Angiogenesis, inflammation, detoxification, glycosylation, one-carbon transfer, apoptosis, cell cycle regulation, cellular senescence. Angiogenesis polymorphisms are reported in Table 4. g Baseline populations are virtually identical between this and a corresponding study reported in Supplementary Table 1. h For stage III and ER negative tumors.
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Tamez S [61] (case series)
TP53 Arg/Arg was associated with better PFS (p = 0.01) NS TP53 Pro/Pro vs Arg/Arg HR 6.4 (1.1–6.4); global p = 0.03 TP 53 Arg/Pro vs Arg/Arg HR 1.7 (0.8–3.7) MDM2 309G/- vs T/T Median OS: 69 vs 34 mos; p = 0.02h VDR FokI C/C vs T/HR 0.2, CI 0.05–0.6, p = 0.006 Shorter AR (CAG ≤19) HR 3.5 (3.0–4.1); p = 0.02 Median PFS: 6 vs 19 mos; p = 0.0001 Longer AR (CAG ≥23) HR 0.5 (0.2–0.9); p = 0.03 All comparisons, p = NS
361
362
Table 4 Ovarian cancer: VEGF polymorphisms and association with outcome. Author (study type) stage Country Prognostic Hefler LAa [72] (case studies series) all stages
Polterauer Sa [74] (case series) all stages
Goode EL series)
b
[48] (case
Predictive studies
Polymorphism
Out- Reported association come
Covariates Additional comments considered
Austria/ 563 Platinum-taxane Germany
VEGF VEGF
− 634G>C − 1154G>A
OS
Simultaneous carriage of homozygous genotypes (634C/C, -1154G/G, -2578C/C) vs others: HR 2.3 (1.3–4.2); p = 0.006
Austria/ 553 Platinum-taxane Germany
VEGF VEGF VEGF
− 2578C>A 405G>C − 460C>T
OS
NS
OS OS
VEGFC rs17697305 HR 1.9 (1.1–3.1); p = 0.01 VEGFC rs1485766 HR 0.7 (0.6-0.9); p = 0.01
Age Tumor grade FIGO stage Age Tumor grade FIGO stage Age Tumor grade FIGO stage Histology Residual disease Age
Australia
Treatment
325 Platinum-taxane
319 No details
VEGF 936C>T VEGFCVEGFC rs17697305 rs1485766
VEGF
405G>C 3 TagSNPc: rs833068 G>A rs3025033 A>G rs2146323 C>A
VEGF VEGF VEGF VEGF
405G>C − 460C>T
OS
Simultaneous carriage of heterozygous genotypes is associated with better OS: HR 0.5 (0.3-0.7); p = 0.001
− 2578C>A − 1154G>/A
PFS
VEGF Haplotypes All others vs AGCGC: HR 1.9,CI 1.04-3.46; p = 0.04
3′UTR C>T -634G>C 936C>T 3′UTR T>A (rs12411) AC repeat + 4422
PFS PFS RR
− 2578C>A − 1154G>/A -460T>C 405G>C 936C>T
RR, PFS, OS
NRP1 3′UTR C>TT/T vs C/C: HR 2.4; p = 0.05 NRP1 3′UTR C>TC/T vs C/C: HR 2.3; p = 0.05 NRP1 3′UTR C>T P = NS for RR All other comparisons (PFS, RR) for VEGF pathway polymorphisms, p = NS All comparisons, p = NS
VEGF
Smerdel MP [76] (case series) stages II–IV;
Denmark 159 Platinum-taxane
Steffensen KD [77] (case series) all stages
Denmark 143 PlatinumVEGF cyclophosphamide VEGF
d
Schultheis AM [59] (phase II clinical trial) relapsed disease
USA and Canada
Smerdel MP [75] (case series) relapsed disease
Denmark 38
53
VEGF VEGF
405G>C -460T>C
VEGF
936C>T
BevacizumabNRP1 cyclophosphamide VEGF VEGF KDR KDR Single-agent VEGF VEGF Bevacizumab VEGF VEGF VEGF
OS
405 CC vs G/- HR 1.9 (1.1-3.2); p = 0.05 rs833068 AA vs G/- HR 2.1 (1.2-3.8); p = 0.05
OS
Tumor grade FIGO stage Histology Age Tumor grade FIGO stage Histology Residual tumor Age Tumor grade FIGO stage Residual disease (1 cm) Haplotype
No association observed between individual VEGF genotypes and OS
Ethnicity Chemotherapy Presurgical CA-125 Location of surgery
These could not be replicated in two independent datasets No associations were observed either for rs3025033 A>G nor rs2146323 C>A Analyses were performed on formalin-fixed, paraffinembedded tumors
No association observed between individual VEGF genotypes and PFS
Platinum sensitivity
No adjustment for multiple comparisons
Not reported by authors
Haplotype analyses did not show any association either
The present table, we only included those polymorphisms directly associated with VEGF pathways. OS, overall survival; PFS, Progression-free Survival; RR, Response, Rate; HR, Hazard Ratio; FIGO, International Federation of Obstetrics and Gynecology; NS, non-significant; VEGF, vascular endothelial growth factor; NRP1, neuropilin-1; KDR, also known as VEGF receptor 2. a Baseline populations are virtually identical between these two studies. b Multiple pathways. Angiogenesis contributed 130 polymorphisms to the analysis, of which VEGF pathway polymorphisms were included; only significant associations are presented. Non-VEGF pathway polymorphisms are reported in Table 3). c TagSNPs utilize linkage disequilibrium to choose relatively independently sorting polymorphisms to represent as markers the genetic heritability of each segment of DNA across the chosen gene. d Other polymorphisms evaluated in Schultheis et al. are reported in Table 3.
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Lose F [73] (population registry) all stages
Gene
USA
N
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Polymorphisms of the VEGF pathway and outcome in ovarian cancer A summary of included studies [48, 59, 72–77] is shown in Table 4. In two studies, haplotypes, but not individual polymorphisms of VEGF were associated with prognosis, [72, 77] while a third study found significance only combining two VEGF polymorphisms [76]. Only two studies described survival associations involving single VEGF polymorphisms, [48, 73] but these prognostic relationships were not seen in other studies. As for VEGF polymorphisms as predictors of benefit from the anti-VEGF antibody, bevacizumab, both studies had fewer than 55 patients, and only a neuropilin-1 (NRP1) polymorphism reached borderline significance [59, 75]. Polymorphisms and Outcome in Endometrial, Cervical, and Vulvar Cancers A summary of included studies [29, 78–88] is shown in Supplementary Table 2. Endometrial cancer studies were generally larger studies, but none adjusted for clinical prognostic factors [78, 81, 88]. Of the cervical and vulvar studies, only two had a sample size greater than 100 patients [89, 83]. Kim et al. reported that VEGF 405C/C (or an associated haplotype) was associated with worse DFS/OS in 199 cervical cancer patients; [83] this finding has not been confirmed independently. Discussion At the international, co-operative group level, the identification of potentially predictive and/or prognostic genetic polymorphisms has been recognized as a high priority. This is the first systematic review of published studies investigating the potential role of genetic polymorphisms as predictive and prognostic biomarkers in gynecological malignancies. The vast majority of pharmacogenomic studies have been undertaken in ovarian cancer. Despite a steady increase in the number of published reports over the past five years, no compelling or consistent association was identified. Instead, there was an absence of uniform, well-defined, and validated putative polymorphic biomarkers that warranted translation into the clinical setting. Meta-analyses or pooled analyses can provide perspective to the individual results. However, when we attempted to perform a meta-analysis of the polymorphisms of the most commonly evaluated genes (ERCC1, VEGF, ABCB1, GSTP1), additional problems surfaced: (i) different genetic models of inheritance and different combinations of genotypes were evaluated across studies, rendering comparability across studies of the currently published literature of limited value; (ii) although techniques such as inverse weighting can be applied, [90] different outcome measure definitions rendered meta-analysis difficult to interpret; (iii) the sample sizes and number of studies reporting negative results for each polymorphism were generally of greater magnitude than the corresponding studies reporting positive results; and (iv) inability to collect unpublished data from individual investigators. Active efforts are required to prospectively coordinate consortia-based formal meta-analyses and pooled analyses of future studies, using standardized variables, definitions, and analytical models. This systematic review has several limitations. The quality of the studies varied, the vast majority of them being either case series or observational studies, and therefore, exploratory in nature. Many studies were underpowered (32% had fewer than 100 patients) to detect any significant association between the polymorphism(s) analyzed and the clinical outcome. Sixteen studies did not perform multivariate analysis, and therefore did not consider the possibility of confounding by other prognostic factors. At least a third of included studies utilized formalin-fixed paraffin embedded specimens for genotyping, without considering whether somatic changes were frequent in the gene regions of the evaluated polymorphisms. Publication bias
363
to positive studies should also be taken into account when interpreting the results of this review. While case series/observational studies did occasionally include sample sizes over 300, patient treatment and follow-up protocols varied substantially. Furthermore, outcome measures were often not uniformly defined across studies (e.g., progressive disease, specific toxicities). This is particularly relevant in ovarian cancer, where the definition of progressive disease upon completion of the primary treatment may vary depending on the criteria used (radiological, clinical, or based on CA-125 changes). Although case–control studies with secondary outcome collection can lead to reasonable analyses of clinical utility (valuable for general prognosis), such analyses may have limitations in the ability to answer key pharmacogenomic evaluations, particularly of drug toxicity. Future observational studies need to report results according to REMARK guidelines [91]. Only during the systematic review process did we discover examples of significant methodological deviations from guidelines; [91] examples include: (i) publication of two parallel analyses of different sets of VEGF polymorphisms involving virtually identical patient populations, using identical outcomes in two separate journals in the same month; appropriate analysis should have considered all evaluated VEGF polymorphisms in a single analysis; [72, 74] (ii) not reporting or acknowledging that a prognostic association was based on only two individuals in one arm, where the first death occurred perioperatively; [14] and (iii) not reporting significant deviation from Hardy Weinberg Equilibrium, especially when genotyping from fixed tissues, raising questions about technical problems with the genotyping process [86]. Few phase III clinical trials have systematically evaluated the relationship between genetic polymorphisms and clinical outcome in gynecologic cancers [22]. Since the completion of HapMap Project, coupled with a rapid progress in genotyping techniques, researchers are now able to investigate much larger numbers of polymorphisms (i.e., up to 2.5 million) at reasonable costs. Comprehensive discovery approaches (e.g., genome-wide association studies) focused on large phase III clinical trials may be required to move the field forward. Large randomized phase III clinical trials can provide the adequate platform for successful genotype–phenotype association-based discovery, but only when the treatment arms represent reasonable therapeutic options. In addition to discovery studies, equal consideration should be given to the publication of high quality adequately-powered validation studies. There is currently a great opportunity and need in gynecologic oncology to implement well-conducted pharmacogenomic studies. This is particularly apparent in epithelial ovarian cancer. The emergence of many promising agents (e.g. poly-ADP ribose polymerase inhibitors), [92–94] and the recent presentation of three studies [95–97] investigating the addition of the anti-angiogenic agent bevacizumab to standard chemotherapy highlight the need for predictive biomarkers to assist clinicians and patients in their choice of treatment. Since biological agents, such as bevacizumab, may require prolonged administration, often until progression, improving therapeutic ratios by finding effective biomarkers would be both a cost-effective and a clinically effective strategy. This systematic review highlights the need for future studies to adhere to the highest methodological standards. Only with large clinical trial samples or carefully annotated prospective observational studies will we be able to conduct adequately powered pre-defined subgroup analysis. In addition, validation of potential useful biomarkers across studies, and even between tumor types, may be a key in identifying clinically useful pharmacogenomic markers. Supplementary materials related to this article can be found online at doi:10.1016/j.ygyno.2011.10.034. Conflict of interest statement The authors declare no conflict of interest for the present manuscript. This study has not been presented in part or in whole elsewhere.
364
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
Acknowledgments The authors want to thank the library research assistance of Rouhi Fazelzad References [1] Siegel R, Ward E, Brawley O, Jemal A. Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 2011;61:212–36. [2] Cannistra SA. Cancer of the ovary. N Engl J Med 2004;351:2519–29. [3] Pectasides D, Pectasides E, Economopoulos T. Systemic therapy in metastatic or recurrent endometrial cancer. Cancer Treat Rev 2007;33:177–90. [4] Pectasides D, Kamposioras K, Papaxoinis G, Pectasides E. Chemotherapy for recurrent cervical cancer. Cancer Treat Rev 2008;34:603–13. [5] Zhu X, Wu S, Dahut WL, Parikh CR. Risks of proteinuria and hypertension with bevacizumab, an antibody against vascular endothelial growth factor: systematic review and meta-analysis. Am J Kidney Dis 2007;49:186–93. [6] Savas S, Liu G. Studying genetic variations in cancer prognosis (and risk): a primer for clinicians. Oncologist 2009;14:657–66. [7] du Bois A, Luck HJ, Meier W, Adams HP, Mobus V, Costa S, et al. A randomized clinical trial of cisplatin/paclitaxel versus carboplatin/paclitaxel as first-line treatment of ovarian cancer. J Natl Cancer Inst 2003;95:1320–9. [8] Secord AA, Havrilesky LJ, O'Malley DM, Bae-Jump V, Fleming ND, Broadwater G, et al. A multicenter evaluation of sequential multimodality therapy and clinical outcome for the treatment of advanced endometrial cancer. Gynecol Oncol 2009;114:442–7. [9] Monk BJ, Sill MW, McMeekin DS, Cohn DE, Ramondetta LM, Boardman CH, et al. Phase III trial of four cisplatin-containing doublet combinations in stage IVB, recurrent, or persistent cervical carcinoma: a Gynecologic Oncology Group study. J Clin Oncol 2009;27:4649–55. [10] Higgins JPT. Chapter 8: Assessing risk of bias in included studies. In: Higgins JPT, editor. Cochrane handbook for systematic reviews of interventions version 5.0.0. [updated February 2008]: The Cochrane Collaboration, 2008. Available: http:// www.cochrane-handbook.org/. [11] Beeghly A, Katsaros D, Chen H, Fracchioli S, Zhang Y, Massobrio M, et al. Glutathione S-transferase polymorphisms and ovarian cancer treatment and survival. Gynecol Oncol 2006;100:330–7. [12] Gadducci A, Di Cristofano C, Zavaglia M, Giusti L, Menicagli M, Cosio S, et al. P53 gene status in patients with advanced serous epithelial ovarian cancer in relation to response to paclitaxel- plus platinum-based chemotherapy and long-term clinical outcome. Anticancer Res 2006;26:687–93. [13] Green H, Soderkvist P, Rosenberg P, Horvath G, Peterson C. mdr-1 single nucleotide polymorphisms in ovarian cancer tissue: G2677T/A correlates with response to paclitaxel chemotherapy. Clin Cancer Res 2006;12:854–9. [14] Green H, Soderkvist P, Rosenberg P, Horvath G, Peterson C. ABCB1 G1199A polymorphism and ovarian cancer response to paclitaxel. J Pharm Sci 2008;97: 2045–8. [15] Heubner M, Wimberger P, Riemann K, Kasimir-Bauer S, Otterbach F, Kimmig R, et al. The CYP1A1 Ile462Val polymorphism and platinum resistance of epithelial ovarian neoplasms. Oncol Res 2010;18:343–7. [16] Johnatty SE, Beesley J, Paul J, Fereday S, Spurdle AB, Webb PM, et al. ABCB1 (MDR 1) polymorphisms and progression-free survival among women with ovarian cancer following paclitaxel/carboplatin chemotherapy. Clin Cancer Res 2008;14: 5594–601. [17] Kang S, Ju W, Kim JW, Park NH, Song YS, Kim SC, et al. Association between excision repair cross-complementation group 1 polymorphism and clinical outcome of platinum-based chemotherapy in patients with epithelial ovarian cancer. Exp Mol Med 2006;38:320–4. [18] Khrunin AV, Moisseev A, Gorbunova V, Limborska S. Genetic polymorphisms and the efficacy and toxicity of cisplatin-based chemotherapy in ovarian cancer patients. Pharmacogenomics J 2010;10:54–61. [19] Kim HS, Kim MK, Chung HH, Kim JW, Park NH, Song YS, et al. Genetic polymorphisms affecting clinical outcomes in epithelial ovarian cancer patients treated with taxanes and platinum compounds: a Korean population-based study. Gynecol Oncol 2009;113:264–9. [20] Krivak TC, Darcy KM, Tian C, Armstrong D, Baysal BE, Gallion H, et al. Relationship between ERCC1 polymorphisms, disease progression, and survival in the Gynecologic Oncology Group Phase III Trial of intraperitoneal versus intravenous cisplatin and paclitaxel for stage III epithelial ovarian cancer. J Clin Oncol 2008;26:3598–606. [21] Krivak TC, Darcy KM, Tian C, Bookman M, Gallion H, Ambrosone CB, et al. Single nucleotide polypmorphisms in ERCC1 are associated with disease progression, and survival in patients with advanced stage ovarian and primary peritoneal carcinoma; a Gynecologic Oncology Group study. Gynecol Oncol 2011;122:121–6. [22] Marsh S, Paul J, King CR, Gifford G, McLeod HL, Brown R. Pharmacogenetic assessment of toxicity and outcome after platinum plus taxane chemotherapy in ovarian cancer: the Scottish Randomised Trial in Ovarian Cancer. J Clin Oncol 2007;25: 4528–35. [23] Morari EC, Lima AB, Bufalo NE, Leite JL, Granja F, Ward LS. Role of glutathione-Stransferase and codon 72 of P53 genotypes in epithelial ovarian cancer patients. J Cancer Res Clin Oncol 2006;132:521–8. [24] Nagle CM, Chenevix-Trench G, Spurdle AB, Webb PM. The role of glutathione-Stransferase polymorphisms in ovarian cancer survival. Eur J Cancer 2007;43: 283–90.
[25] Obata H, Yahata T, Quan J, Sekine M, Tanaka K. Association between single nucleotide polymorphisms of drug resistance-associated genes and response to chemotherapy in advanced ovarian cancer. Anticancer Res 2006;26:2227–32. [26] Saldivar JS, Lu KH, Liang D, Gu J, Huang M, Vlastos AT, et al. Moving toward individualized therapy based on NER polymorphisms that predict platinum sensitivity in ovarian cancer patients. Gynecol Oncol 2007;107:S223–9. [27] Smith S, Su D. Rigault de la Longrais IA, Schwartz P, Puopolo M, Rutherford TJ, Mor G, Yu H, Katsaros D. ERCC1 genotype and phenotype in epithelial ovarian cancer identify patients likely to benefit from paclitaxel treatment in addition to platinum-based therapy. J Clin Oncol 2007;25:5172–9. [28] Steffensen KD, Waldstrom M, Jeppesen U, Brandslund I, Jakobsen A. Prediction of response to chemotherapy by ERCC1 immunohistochemistry and ERCC1 polymorphism in ovarian cancer. Int J Gynecol Cancer 2008;18:702–10. [29] Takano M, Kato M, Yoshikawa T, Sasaki N, Hirata J, Furuya K, et al. Clinical significance of UDP-glucuronosyltransferase 1A1*6 for toxicities of combination chemotherapy with irinotecan and cisplatin in gynecologic cancers: a prospective multi-institutional study. Oncology 2009;76:315–21. [30] Germann UA. P-glycoprotein–a mediator of multidrug resistance in tumour cells. Eur J Cancer 1996;32A:927–44. [31] Hoffmeyer S, Burk O, von Richter O, Arnold HP, Brockmoller J, Johne A, et al. Functional polymorphisms of the human multidrug-resistance gene: multiple sequence variations and correlation of one allele with P-glycoprotein expression and activity in vivo. Proc Natl Acad Sci U S A 2000;97:3473–8. [32] Baekelandt MM, Holm R, Nesland JM, Trope CG, Kristensen GB. P-glycoprotein expression is a marker for chemotherapy resistance and prognosis in advanced ovarian cancer. Anticancer Res 2000;20:1061–7. [33] Yu JJ, Lee KB, Mu C, Li Q, Abernathy TV, Bostick-Bruton F, et al. Comparison of two human ovarian carcinoma cell lines (A2780/CP70 and MCAS) that are equally resistant to platinum, but differ at codon 118 of the ERCC1 gene. Int J Oncol 2000;16:555–60. [34] Quintela-Fandino M, Hitt R, Medina PP, Gamarra S, Manso L, Cortes-Funes H, et al. DNA-repair gene polymorphisms predict favorable clinical outcome among patients with advanced squamous cell carcinoma of the head and neck treated with cisplatin-based induction chemotherapy. J Clin Oncol 2006;24:4333–9. [35] Zhou W, Gurubhagavatula S, Liu G, Park S, Neuberg DS, Wain JC, et al. Excision repair cross-complementation group 1 polymorphism predicts overall survival in advanced non-small cell lung cancer patients treated with platinum-based chemotherapy. Clin Cancer Res 2004;10:4939–43. [36] Strange RC, Spiteri MA, Ramachandran S, Fryer AA. Glutathione-S-transferase family of enzymes. Mutat Res 2001;482:21–6. [37] McIlwain CC, Townsend DM, Tew KD. Glutathione S-transferase polymorphisms: cancer incidence and therapy. Oncogene 2006;25:1639–48. [38] Batra J, Tan OL, O'Mara T, Zammit R, Nagle CM, Clements JA, et al. Kallikrein-related peptidase 10 (KLK10) expression and single nucleotide polymorphisms in ovarian cancer survival. Int J Gynecol Cancer 2010;20:529–36. [39] Galic V, Willner J, Wollan M, Garg R, Garcia R, Goff BA, et al. Common polymorphisms in TP53 and MDM2 and the relationship to TP53 mutations and clinical outcomes in women with ovarian and peritoneal carcinomas. Genes Chromosomes Cancer 2007;46:239–47. [40] Grimm C, Polterauer S, Zeillinger R, Tempfer C, Sliutz G, Reinthaller A, et al. The prohibitin 3′ untranslated region polymorphism in patients with ovarian cancer. Eur J Obstet Gynecol Reprod Biol 2008;137:236–9. [41] Grimm C, Polterauer S, Zeillinger R, Tong D, Heinze G, Wolf A, et al. Two multidrug-resistance (ABCB1) gene polymorphisms as prognostic parameters in women with ovarian cancer. Anticancer Res 2010;30:3487–91. [42] Ioana Braicu E, Mustea A, Toliat MR, Pirvulescu C, Konsgen D, Sun P, et al. Polymorphism of IL-1alpha, IL-1beta and IL-10 in patients with advanced ovarian cancer: results of a prospective study with 147 patients. Gynecol Oncol 2007;104:680–5. [43] Mann A, Hogdall E, Ramus SJ, DiCioccio RA, Hogdall C, Quaye L, et al. Mismatch repair gene polymorphisms and survival in invasive ovarian cancer patients. Eur J Cancer 2008;44:2259–65. [44] Song H, Hogdall E, Ramus SJ, Dicioccio RA, Hogdall C, Quaye L, et al. Effects of common germ-line genetic variation in cell cycle genes on ovarian cancer survival. Clin Cancer Res 2008;14:1090–5. [45] Bartel F, Jung J, Bohnke A, Gradhand E, Zeng K, Thomssen C, et al. Both germ line and somatic genetics of the p53 pathway affect ovarian cancer incidence and survival. Clin Cancer Res 2008;14:89–96. [46] Batra J, Nagle CM, O'Mara T, Higgins M, Dong Y, Tan OL, et al. A Kallikrein 15 (KLK15) single nucleotide polymorphism located close to a novel exon shows evidence of association with poor ovarian cancer survival. BMC Cancer 2011;11:119. [47] Garg R, Wollan M, Galic V, Garcia R, Goff BA, Gray HJ, et al. Common polymorphism in interleukin 6 influences survival of women with ovarian and peritoneal carcinoma. Gynecol Oncol 2006;103:793–6. [48] Goode EL, Maurer MJ, Sellers TA, Phelan CM, Kalli KR, Fridley BL, et al. Inherited determinants of ovarian cancer survival. Clin Cancer Res 2010;16:995–1007. [49] Han CH, Huang YJ, Lu KH, Liu Z, Mills GB, Wei Q, et al. Polymorphisms in the SULF1 gene are associated with early age of onset and survival of ovarian cancer. J Exp Clin Cancer Res 2011;30:5. [50] Hefler LA, Grimm C, Ackermann S, Malur S, Radjabi-Rahat AR, Leodolter S, et al. An interleukin-6 gene promoter polymorphism influences the biological phenotype of ovarian cancer. Cancer Res 2003;63:3066–8. [51] Higashi T, Kyo S, Inoue M, Tanii H, Saijoh K. Novel functional single nucleotide polymorphisms in the latent transforming growth factor-beta binding protein1L promoter: effect on latent transforming growth factor-beta binding protein-
I. Diaz-Padilla et al. / Gynecologic Oncology 124 (2012) 354–365
[52] [53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64] [65] [66]
[67]
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
1L expression level and possible prognostic significance in ovarian cancer. J Mol Diagn 2006;8:342–50. Li AJ, Baldwin RL, Karlan BY. Short androgen receptor allele length is a poor prognostic factor in epithelial ovarian carcinoma. Clin Cancer Res 2003;9:3667–73. Liang D, Meyer L, Chang DW, Lin J, Pu X, Ye Y, et al. Genetic variants in MicroRNA biosynthesis pathways and binding sites modify ovarian cancer risk, survival, and treatment response. Cancer Res 2010;70:9765–76. Ludwig AH, Murawska M, Panek G, Timorek A, Kupryjanczyk J. Androgen, progesterone, and FSH receptor polymorphisms in ovarian cancer risk and outcome. Endocr Relat Cancer 2009;16:1005–16. Nagle CM, Chenevix-Trench G, Webb PM, Spurdle AB. Ovarian cancer survival and polymorphisms in hormone and DNA repair pathway genes. Cancer Lett 2007;251:96–104. Paige AJ, Zucknick M, Janczar S, Paul J, Mein CA, Taylor KJ, et al. WWOX tumour suppressor gene polymorphisms and ovarian cancer pathology and prognosis. Eur J Cancer 2010;46:818–25. Quaye L, Gayther SA, Ramus SJ, Di Cioccio RA, McGuire V, Hogdall E, et al. The effects of common genetic variants in oncogenes on ovarian cancer survival. Clin Cancer Res 2008;14:5833–9. Santos AM, Sousa H, Portela C, Pereira D, Pinto D, Catarino R, et al. TP53 and P21 polymorphisms: response to cisplatinum/paclitaxel-based chemotherapy in ovarian cancer. Biochem Biophys Res Commun 2006;340:256–62. Schultheis AM, Lurje G, Rhodes KE, Zhang W, Yang D, Garcia AA, et al. Polymorphisms and clinical outcome in recurrent ovarian cancer treated with cyclophosphamide and bevacizumab. Clin Cancer Res 2008;14:7554–63. Six L, Grimm C, Leodolter S, Tempfer C, Zeillinger R, Sliutz G, et al. A polymorphism in the matrix metalloproteinase-1 gene promoter is associated with the prognosis of patients with ovarian cancer. Gynecol Oncol 2006;100:506–10. Tamez S, Norizoe C, Ochiai K, Takahashi D, Shimojima A, Tsutsumi Y, et al. Vitamin D receptor polymorphisms and prognosis of patients with epithelial ovarian cancer. Br J Cancer 2009;101:1957–60. Wynendaele J, Bohnke A, Leucci E, Nielsen SJ, Lambertz I, Hammer S, et al. An illegitimate microRNA target site within the 3′UTR of MDM4 affects ovarian cancer progression and chemosensitivity. Cancer Res 2010;70:9641–9. Reles A, Wen WH, Schmider A, Gee C, Runnebaum IB, Kilian U, et al. Correlation of p53 mutations with resistance to platinum-based chemotherapy and shortened survival in ovarian cancer. Clin Cancer Res 2001;7:2984–97. Pietsch EC, Humbey O, Murphy ME. Polymorphisms in the p53 pathway. Oncogene 2006;25:1602–11. Bond GL, Hu W, Levine AJ. MDM2 is a central node in the p53 pathway: 12 years and counting. Curr Cancer Drug Targets 2005;5:3–8. Dijsselbloem N, Goriely S, Albarani V, Gerlo S, Francoz S, Marine JC, et al. A critical role for p53 in the control of NF-kappaB-dependent gene expression in TLR4stimulated dendritic cells exposed to Genistein. J Immunol 2007;178:5048–57. Phelps M, Darley M, Primrose JN, Blaydes JP. p53-independent activation of the hdm2-P2 promoter through multiple transcription factor response elements results in elevated hdm2 expression in estrogen receptor alpha-positive breast cancer cells. Cancer Res 2003;63:2616–23. Risch HA. Hormonal etiology of epithelial ovarian cancer, with a hypothesis concerning the role of androgens and progesterone. J Natl Cancer Inst 1998;90: 1774–86. Evangelou A, Jindal SK, Brown TJ, Letarte M. Down-regulation of transforming growth factor beta receptors by androgen in ovarian cancer cells. Cancer Res 2000;60:929–35. Cardillo MR, Petrangeli E, Aliotta N, Salvatori L, Ravenna L, Chang C, et al. Androgen receptors in ovarian tumors: correlation with oestrogen and progesterone receptors in an immunohistochemical and semiquantitative image analysis study. J Exp Clin Cancer Res 1998;17:231–7. Buchanan G, Yang M, Cheong A, Harris JM, Irvine RA, Lambert PF, et al. Structural and functional consequences of glutamine tract variation in the androgen receptor. Hum Mol Genet 2004;13:1677–92. Hefler LA, Mustea A, Konsgen D, Concin N, Tanner B, Strick R, et al. Vascular endothelial growth factor gene polymorphisms are associated with prognosis in ovarian cancer. Clin Cancer Res 2007;13:898–901. Lose F, Nagle CM, O'Mara T, Batra J, Bolton KL, Song H, et al. Vascular endothelial growth factor gene polymorphisms and ovarian cancer survival. Gynecol Oncol 2010;119:479–83. Polterauer S, Grimm C, Mustea A, Concin N, Tanner B, Thiel F, et al. Vascular endothelial growth factor gene polymorphisms in ovarian cancer. Gynecol Oncol 2007;105:385–9. Smerdel MP, Steffensen KD, Waldstrom M, Brandslund I, Jakobsen A. The predictive value of serum VEGF in multiresistant ovarian cancer patients treated with bevacizumab. Gynecol Oncol 2010;118:167–71. Smerdel MP, Waldstrom M, Brandslund I, Steffensen KD, Andersen RF, Jakobsen A. Prognostic importance of vascular endothelial growth factor-A expression and
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
365
vascular endothelial growth factor polymorphisms in epithelial ovarian cancer. Int J Gynecol Cancer 2009;19:578–84. Steffensen KD, Waldstrom M, Brandslund I, Jakobsen A. The relationship of VEGF polymorphisms with serum VEGF levels and progression-free survival in patients with epithelial ovarian cancer. Gynecol Oncol 2010;117:109–16. Amano M, Yoshida S, Kennedy S, Takemura N, Deguchi M, Ohara N, et al. Association study of vascular endothelial growth factor gene polymorphisms in endometrial carcinomas in a Japanese population. Eur J Gynaecol Oncol 2008;29:333–7. Cheng XD, Lu WG, Ye F, Wan XY, Xie X. The association of XRCC1 gene single nucleotide polymorphisms with response to neoadjuvant chemotherapy in locally advanced cervical carcinoma. J Exp Clin Cancer Res 2009;28:91. Chung HH, Kim MK, Kim JW, Park NH, Song YS, Kang SB, et al. XRCC1 R399Q polymorphism is associated with response to platinum-based neoadjuvant chemotherapy in bulky cervical cancer. Gynecol Oncol 2006;103:1031–7. Einarsdottir K, Darabi H, Czene K, Li Y, Low YL, Li YQ, et al. Common genetic variability in ESR1 and EGF in relation to endometrial cancer risk and survival. Br J Cancer 2009;100:1358–64. Kim K, Kang SB, Chung HH, Kim JW, Park NH, Song YS. XRCC1 Arginine194Tryptophan and GGH-401Cytosine/Thymine polymorphisms are associated with response to platinum-based neoadjuvant chemotherapy in cervical cancer. Gynecol Oncol 2008;111:509–15. Kim YH, Kim MA, Park IA, Park WY, Kim JW, Kim SC, et al. VEGF polymorphisms in early cervical cancer susceptibility, angiogenesis, and survival. Gynecol Oncol 2010;119:232–6. Kim YH, Park IA, Park WY, Kim JW, Kim SC, Park NH, et al. Hypoxia-inducible factor 1alpha polymorphisms and early-stage cervical cancer. Int J Gynecol Cancer 2011;21:2–7. Lerma E, Romero M, Gallardo A, Pons C, Munoz J, Fuentes J, et al. Prognostic significance of the Fas-receptor/Fas-ligand system in cervical squamous cell carcinoma. Virchows Arch 2008;452:65–74. Mehta AM, Jordanova ES, Corver WE, van Wezel T, Uh HW, Kenter GG, et al. Single nucleotide polymorphisms in antigen processing machinery component ERAP1 significantly associate with clinical outcome in cervical carcinoma. Genes Chromosomes Cancer 2009;48:410–8. Riener EK, Hefler LA, Grimm C, Galid A, Zeillinger R, Tong-Cacsire D, et al. Polymorphisms of the endothelial nitric oxide synthase gene in women with vulvar cancer. Gynecol Oncol 2004;93:686–90. Rodriguez G, Bilbao C, Ramirez R, Falcon O, Leon L, Chirino R, et al. Alleles with short CAG and GGN repeats in the androgen receptor gene are associated with benign endometrial cancer. Int J Cancer 2006;118:1420–5. Kim YH, Park I-A, Park W-Y, Kim JW, Kim SC, Park N-H, et al. Hypoxia-inducible factor 1 polymorphisms and early-stage cervical cancer. Int J Gynecol Cancer 2011;21:2–7. Cronin-Fenton DP, Lash TL. Clinical epidemiology and pharmacology of CYP2D6 inhibition related to breast cancer outcomes. Expert Rev Clin Pharmacol 2011;4: 363–77. McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM. Reporting recommendations for tumor marker prognostic studies (REMARK). J Natl Cancer Inst 2005;97:1180–4. Ledermann JA, Harter P, Gourley C, Friedlander M, Vergote IB, Rustin GJS, et al. Phase II randomized placebo-controlled study of olaparib (AZD2281) in patients with platinum-sensitive relapsed serous ovarian cancer (PSR SOC). J Clin Oncol 2011;29 (suppl; abstr 5003). Birrer MJ, Konstantinopoulos P, Penson RT, Roche M, Ambrosio A, Stallings TE, et al. A phase II trial of iniparib (BSI-201) in combination with gemcitabine/ carboplatin (GC) in patients with platinum-resistant recurrent ovarian cancer. J Clin Oncol 2011;29 (suppl; abstr 5005). Penson RT, Whalen C, Lasonde B, Krasner CN, Konstantinopoulos P, Stallings TE, et al. A phase II trial of iniparib (BSI-201) in combination with gemcitabine/carboplatin (GC) in patients with platinum-sensitive recurrent ovarian cancer. J Clin Oncol 2011;29 (suppl; abstr 5004). Burger RA, Brady MF, Bookman MA, Walker JL, Homesley HD, Fowler J, et al. Phase III trial of bevacizumab (BEV) in the primary treatment of advanced epithelial ovarian cancer (EOC), primary peritoneal cancer (PPC), or fallopian tube cancer (FTC): A Gynecologic Oncology Group study. J Clin Oncol 2010;28(18s) (suppl; abstr LBA1). Kristensen G, Perren T, Qian W, Pfisterer J, Ledermann JA, Joly F, et al. Result of interim analysis of overall survival in the GCIG ICON7 phase III randomized trial of bevacizumab in women with newly diagnosed ovarian cancer. J Clin Oncol 2011;29 (suppl; abstr LBA5006). Aghajanian C, Finkler NJ, Rutherford T, Smith DA, Yi J, Parmar H, et al. OCEANS: a randomized, double-blinded, placebo-controlled phase III trial of chemotherapy with or without bevacizumab (BEV) in patients with platinum-sensitive recurrent epithelial ovarian (EOC), primary peritoneal (PPC), or fallopian tube cancer (FTC). J Clin Oncol 2011;29 (suppl; abstr LBA5007).