Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Jun 1.
Published in final edited form as: Gynecol Oncol. 2010 Mar 16;117(3):429–439. doi: 10.1016/j.ygyno.2010.01.048

Translational Research in the Gynecologic Oncology Group: Evaluation of Ovarian Cancer Markers, Profiles and Novel Therapies

Kathleen M Darcy 1, Michael J Birrer 2
PMCID: PMC2877379  NIHMSID: NIHMS180250  PMID: 20233625

Abstract

Objectives

To review the translational research (TR) performed in the Gynecologic Oncology Group (GOG) to evaluate ovarian cancer markers, profiles and novel therapies.

Methods

Prospective trials with stand alone or embedded TR objectives involving patient and specimen accrual as well as retrospective studies using banked specimens and resources were and continue to be performed in the GOG. Appropriate statistical methods are employed to evaluate associations with clinical characteristics and outcomes including tumor response, adverse events, progression free survival and overall survival.

Results

Highlights are presented for some of the collaborative and multidisciplinary TR conducted with the GOG to evaluate markers, pathway and novel therapeutics in epithelial ovarian, primary peritoneal and/or fallopian tube cancer. For example, in GOG 111, high immunohistochemical (IHC) expression of cyclin E was associated with a shorter median survival (29 versus 35 months) and an increased risk of death (hazard ratio [HR]=1.4, 95% confidence interval [CI]=1.0–2.1, p=0.05). In GOG 114/132, non-detectable immunoblot expression of maspin was associated with debulking status (p=0.034) and an increased risk of disease progression (HR=1.89, 95% CI=1.04–3.45, p=0.038) and death (HR=1.99, 95% CI=1.07–3.69, p=0.030) while high CD105-microvessel density (MVD), but not CD31-MVD in tumor was associated with increased risk of disease progression (HR=1.873, 95% CI=1.102–3.184, p=0.020) but not death. In GOG 172, low IHC expression of BRCA1 was associated with advanced stage (p<0.001), serous histology (p<0.001) and a reduced risk of disease progression (HR=0.64, 95% CI=0.42–0.96) and death (HR=0.51, 95% CI=0.32–0.83) while the CA/AA versus CC genotypes in C8092A in ERCC1 were associated with an increased risk of disease progression (HR=1.44, 95% CI=1.06–1.94, p=0.018) and death (HR=1.50, 95% CI=1.07–2.09, p=0.018).

Conclusions

The GOG has an extensive TR program that provides clues regarding the molecular and biochemical mechanisms of disease, treatments and outcomes in women with or at risk for a gynecologic malignancy.

Keywords: Ovarian Cancer, Markers, Profiles, Tumor Biology, Novel Therapies, Translational Research

INTRODUCTION

Translational research (TR) is the bridge between clinical research and basic science that provides clues regarding the molecular and biochemical mechanisms of disease, treatments and outcomes in clinical trials, and the rationale for integrating advances in oncology, science, technologies and drug development into clinical trials and practice. Given the plethora of agents and modalities available for testing in clinical trials, coordinated and collaborative approaches are needed to move the most promising regimens through the drug development process as rapidly and efficiently as possible. Issues like costs, reimbursements and access must also be considered during the drug development process as it is not sufficient to define new standards of care if the more effective treatments can not be adopted into the continuum of clinical practices in the community.

The Gynecologic Oncology Group (GOG) is a multidisciplinary and international Cooperative Trial Group with a TR program that evaluates markers and profiles with potential diagnostic, prognostic and predictive value in prospective trials involving patient and specimen accrual as well as retrospective studies using banked specimens and resources. The GOG recognizes that TR is a critical element of cooperative group clinical trials in the 21st century and the success of these studies requires the integration of objectives that are scientifically-sound and hypothesis-based; standard operating procedures and training that permit member institutions to submit high quality data and specimens; experienced and funded laboratories with appropriate expertise and validated conventional and high through-put assays; and an infrastructure with well-annotated specimens and resources for cutting-edge TR that improves clinical management, outcomes and quality of life. This review starts with overviews about cancer biology, signal transduction and cancer therapeutics and then provides highlights of some of the collaborative and multidisciplinary TR conducted with the GOG to evaluate markers, pathway and novel therapeutics in ovarian, primary peritoneal and/or Fallopian tube cancer. Ultimately, GOG phase II and III trials will selectively treat and manage patients based on markers and profiles, and personalized medicine will become the new standard of care for women with gynecologic malignancies.

Cancer Biology

The Cancer Genome Atlas (TCGA) project in ovarian cancer is beginning to release data that is confirming that epithelial ovarian cancers (EOCs) exhibit extensive molecular heterogeneity with alterations in numerous pathways including oncogenes, tumor suppressors, cell cycle regulation and DNA repair. EOCs exhibit aneuploidy, chromosomal alterations, genomic instability, mutations, amplifications, overepxression, amplifications, silencing, modifications, splicing and epigenetic mechanisms as well as natural and induced sequence variations. Genomic and epigenetic alterations not only drive tumorigenesis, invasion, metastasis and disease progression, but also affect which patients will or won’t respond to specific treatments or experience adverse events. Among the hundreds of defects and alterations observed within the tumors in individual patients with EOC, most are likely passengers while only a fraction are species-specific operators (drivers). Identification of the casual drivers in individual EOC patients will enable us to design more effective marker-driven clinical trials that select the right drugs for the right patients.

Although the molecular classification of EOC clearly represents a landmark advance for women with a diagnosis of EOC or increased risk of this disease, the more challenging work lies before us. Effective treatments with long-term clinical benefit will not only require sustained inactivation or re-control of the critical drivers of tumorigenesis operating in a particular cancer patient but must also anticipate and counteract natural feedback loops, redundant and divergent genes and pathways as well as innate and acquired resistance mechanisms that are differentially induced in select EOC patients. This includes drug efflux, metabolism, detoxification, clearance along with DNA repair pathways, expression, post-translational modifications, silencing, alternative splicing, isoform-switching and epithelial-mesenchymal transitioning, for example. It is also becoming clear that a number of the molecular defects and mechanisms operative in EOC vary more by cell type and grade than by disease site. A number of clinical trials are already focusing eligibility criteria on select histologies within or across disease sites.

Signal Transduction

Tumorigenesis, invasion, metastasis and disease progression can be controlled of membrane-bound, cytoplasmic and nuclear receptors that can be activated by ligands. Following activation, receptors dimerize or oligiomerize and undergo conformational changes, autophosphorylation and phosphorylation of signaling molecules that ultimately regulate transcription, translation, and post-translational modifications as well as processes affecting cell proliferation, maturation, contact, adhesion, migration, invasion, survival, resistance, and the production and secretion of growth factors, cytokines, chemokines and soluble receptors (Figure 1A and 1B). These autocrine, paracrine and systemic factors then affect different cells in the tumor microenvironment and distant sites thus further regulating cancer progression, angiogenesis, vasculogenesis, permeability, immune function as well as the efficacy and toxicities associated with cancer treatments.

Figure 1.

Figure 1

Panel A provides a schematic representation of ErbB receptor family members and ligands (as referenced by Darcy et al. [51,52]). The ErbB receptor family, composed of epidermal growth factor receptor (EGFR)ErbB1/Her2, ErbB2Her2, ErbB3Her3 and ErbB4Her4, has an extracellular ligand binding domain, a transmembrane domain and a cytoplamic tyrosine kinase domain. EGFR ligands include EGF, TGFα, HB-EGF, amphiregulin, betacellulin and epiregulin. ErbB2 has very high intrinsic tyrosine kinase activity in the absence of any known ligands. ErbB3 has a very weak tyrosine kinase domain and is activated by heregulins. NRG2, NRG3, heregulins and betacellulin are ligands for ErbB4. Upon binding ligand, the membrane receptor tyrosine kinases undergo conformation changes, dimerization and autophosphorylation which triggers recruitment of substrates and docking proteins, and phosphorylation of substrates. The distinct ErbB receptor homodimers and heterodimers differentially induce a distinct spectrum of signal transduction cascades and cellular effects. Molecular targeting agents are provided in yellow boxes for EGFR, ErbB2 and various downstream signaling molecules including Raf, MEKMAPKK, Src, PI3K, AKTPKB, mTOR and PKC. Panel B provides a schematic illustration of vascular endothelial growth factor receptor (VEGFR) activation by vascular endothelial growth factor-A (VEGF-A) [53] which induces conformational changes, dimerization and autophosphorylation of this membrane receptor tyrosine kinase. Various signal transduction cascades are induced following activation of the VEGFR which regulate cellular effects including cell proliferation, differentiation, survival, apoptosis, migration, invasion, resistance, permeability, angiogenesis and vasculogenesis. Molecular targeting agents are provided in yellow boxes with lines and a red X to indicate the drug target. Panel C illustrates the functional domains of the wild type p53 tumor suppressor with the binding sites for the N-terminal DO-1 and DO-7 monoclonal antibodies against p53 (as referenced by Darcy et al. [3,13]).

Inappropriate receptor activation promotes tumorigenesis and can be induced by a number of mechanisms including overexpression of autocrine and paracrine factors. Receptors can also be mutated causing constitutive activation in the absence of ligand binding, or be overexpressed via gene amplification, transcriptional activation or post-transcriptional mechanisms which typically require ligand availability and binding for activation. Cross talk between different receptor super families can also activate receptors by a ligand-independent mechanism. Cancer progression, invasion and metastasis are promoted by the amplification, mutation or overexpression of signaling molecules downstream from receptors. Various cell types within the tumor microenvironment can be induced to secrete pro-inflammatory factors that stimulate the vasculature to recruit leukocytes to the tumor. After activation, these tumorassociated leukocytes can release factors that recruit more inflammatory cells and stimulate angiogenesis and neovasculogenesis to sustain tumor growth, promote disease progression, and facilitate tumor invasion and metastasis. Schematics are provided for p53 (Figure 1C), cell cycle regulation (Figure 2A), effects of genotoxic stress (Figure 2B), nucleotide excision repair (Figure 2C) and BRCA1 (Figure 2D) as these pathways are the subject of a number of TR studies conducted by the GOG.

Figure 2.

Figure 2

Panel A depicts some of the regulators of cell cycle progression with emphasis on the transition from G0/G1 to S phase of the cell cycle [54]. Panel B illustrates some of the regulators of cell cycle progression, p53-dependent gene expression, apoptosis and DNA repair following genotoxic stress [55]. Panel C provides a schematic representation and brief description of the steps involved in nucleotide excision repair follow DNA damage [56]. Step 1. DNA is damaged by drugs and radiation. Step 2. XPE-DDB1 complex binds to damaged DNA. Step 3. XPE-DDB1 complex assists in recruiting XPC-HR23B. Step 4. XPC-HR23B complex binds directly to the damaged DNA and XPE-DDB1 complex is released. Step 5. TFIIH complex with XPB and XPD binds to the damaged DNA. Step 6. XPB and XPD helicases unwind the DNA helix. Step 7. XPA, RPA and XPG bind sequentially and the XPC-HR23B complex is released and recycled. Step 8. The ERCC1-XPF exonuclease is recruited. Step 9. Incisions are induced on the 5’ side (ERCC1) and the 3’ side (XPG) of the damaged DNA. Step 10. XPA, TFIIH complex with XPD and XPB, and the oligonucleotide with the damage are removed. Step 11. The resulting gap is filled by a DNA polymerase and RFC, PCNA and ligase I are recruited. XPF-ERCC1 and XPG are displaced. Step 12. DNA is ligated and proteins are released. Panel D illustrates the functional domains within BRCA1 and the general localization of binding sites for a number of the proteins that interact with BRCA1 and are involved in cell cycle regulation, DNA repair and chromatin remodeling [5759].

Cancer Therapeutics

Insights into the molecular and biochemical mechanisms operative in cancer development, progression and metastasis have uncovered a wide array of molecules in tumor cells and/or the tumor microenvironment including stromal cells, endothelial cells, endothelial precursor cells, pericytes, and immune cells that can be targeted therapeutically. Among these agents are the molecular targeting therapies that inhibit receptor tyrosine kinases, non-receptor tyrosine kinases, serine/threonine kinases, transferases, proteases as well as other enzymes, processes and/or pathways. Some of the molecular targeting therapies are selective inhibitors while others are dual inhibitors or multiple inhibitors (Table 1). Figures 1A and 1B provide a few examples of molecular targeting agents that can inhibit epidermal growth factor (EGF), EGF receptor (EGFR)ErbB1/Her1, ErbB2Her2, vascular endothelial growth factor-A (VEGF-A), VEGF receptor (VEGFR) or downstream signaling molecules. A number of these agents are being evaluated in human EOC and specifically in GOG clinical trials as illustrated in Figures 1A and 1B. In addition to the molecular targeting agents, there is an arsenal of traditional cytotoxic anticancer drugs (Table 2). Alkylating agents and microtuble inhibitors have been particular effective in EOC. Despite high initial response rates to first-line treatment and re-challenge with platinum agents and taxanes, about 30% of women with advanced stage EOC fail to respond to initial platinum-taxane based chemotherapy and 5-year survival remains below 40% for women with advanced stage EOC who underwent surgical staging and cytoreduction.

Table 1.

Molecular Targeting Anti-Neoplastic Agents

Mode of Action of Selective Inhibitors Agents
  Inhibit vascular endothelial growth factor (VEGF) bevacizumab VEGF-trap
  Inhibit VEGF receptor (VEGFR) vatalanib cediranib
  Inhibit epidermal growth factor receptor (EGFR) gefitinib
cetuximab
ABX-EGF
erlotinib
matuzumab
  Inhibit human EGF receptor 2 (Her2) / ErbB2 trastuzumab
CP-724,714
SUCI02
  Inhibit MEK / mitogen activated protein kinase
  kinase (MAP2K) / MKK
AZD6244
  Inhibit mammalian target of rapamycin (mTOR) /
  FK506 binding protein 12-rapamycin associated
  protein 1 (FRAP1)
rapamycin
RAD001
temsirolimus
AP23573
  Inhibit protein kinase C (PKC) isoforms brystatin-1 CGP41251
  Inhibit PKC-beta enzastaurin
  Inhibit poly ADP ribose polymerase (PARP) MK-4827
BSI-201
INO-1001
ABT-888
Olaparib
AG140699
  Inhibit ATM LY294002 KU-55933
  Inhibit Notch MK0752
  Inhibit AKT / protein kinase B (PKB) MK-2206 API-59-OME
  Inhibit Src family PD173956
PD180970
PD173958
AP23846
  Inhibit CD105 / endoglin TRC105
  Inhibit hepatocyte growth factor (HGF)/scatter factor AMG 102
  Inhibit Janus kinase 2 (JAK2) cucurbitacin-A
  Inhibit signal transducers and activators of
  transcription 3 (STAT3)
cucurbitacin-Q
  Inhibit phosphoinositide-3 kinase (PI3K) LYC294002
ZSTK474
Wortmannin
  Inhibit Raf CGP 69846A
  Inhibit p38 mitogen-activated protein kinase (MAPK)
  / RK / CSBP
SB203580
  Inhibit Aurora kinases MK-0457
VX-680
L-001281814
  Inhibit cyclin dependent kinase (CDK) pathways flavopiridol
BMS-387032
seliciclib
PD 0332991
  Inhibit hypoxia-inducible factor-1 alpha (HIF-1alpha) EZN-2968
  Inhibit cyclooxygenase-2 (COX-2) celecoxib
rofecoxib
valdecoxib
  Inhibit 26S proteasome bortezomib
  Inhibit farnesyl transferase tipifarnib
BMS-214662
ionafarnib
L778123
  Inhibit matrix metalloproteinases (MMPs) marimastat BAY 12–9566
  Inhibit urokinase-type plasminogen activator (uPA)
  system
WX-UK1
Urokinase-derived
Peptide A6
WX-671
  Inhibit histone deacetylase (HDAC) vorinostat
trichostatin A
valproic acid
LBH589
  Inhibit heat shock protein 90 (HSP90) 17-allylamino-17-
demethoxygeldanamycin
geldanamycin
  Inhibit estrogen receptors (ER) tamoxifen raloxifin
  Inhibit estrogen metabolism letrazole
exemestane
anastrozole
  Inhibit progesterone receptors (PR) provera depoprovera
  Synthetic retinoid fenretinide
Mode of Action of Dual Inhibitors
  Inhibit EGFR and Her2 / ErB2 lapatinab
  Inhibit EGFR and VEGFR ZD6474 AEE788
  Inhibit VEGFR and PDGFR axitinib vandetanib
  Inhibit VEGFR and FGFR brivanib CP-547,632
  Inhibit VEGF and bFGF thalidomide
  Inhibit JAK2 / STAT3 cucurbitacin-B
cucurbitacin-I
cucurbitacin-E
  Inhibit Chk1 and Chk2 AZD-7762 PF-00477736
Mode of Action of Multiple Inhibitors
  Inhibit EGFR, Her2, ErbB3 and ErbB4 canertinib
  Inhibit bcr-abl, c-Kit and PDGFR imatinib
  Inhibit VEGFR, PDGFR and FGFR JNJ-17029259 BIBF1120
  Inhibit VEGFR, PDGFR and Flt3 SU11657 ABT-869
  Inhibit VEGFR, PDGFR and Raf sorafenib
  Inhibit VEGFR, PDGFR and c-Kit sunitinib
AMG 507
BAY 57–9352
AMG-706
AG-013736
  Inhibit VEGFR, PDGFR, c-Kit, CSFR, Flt3 and
  FGFR
CHIR-258
  Inhibit Bcr-Abl, Src and Ephrins dasatinib
  Inhibit Akt, FGFR3 and FLT3 benzoylstaurosporin
  Inhibit VEGFR, PDGFR, CDK1 and CDK2 ZK304709
  Inhibit Raf, VEGFR-2, VEGFR-3, PDGFR-beta,
  FLT3, c-Kit and p38-alpha MAPK
BAY 43–9006
  Inhibit MEK, Erk1 and Erk2 UO126
PD184352
PD98059
  Inhibit PKC, Chk1 and Chk2 UCN-01
Recombinant adenovirus
  Encoding p53 Advexin
SCH58500
Gendicine

Table 2.

Cytotoxic Anti-Neoplastic Agents.

Mode of Action Agents
  Alkylate / cross-link DNA cisplatin
cyclophosphamide
carboplatin
  Inhibit microtubules paclitaxel
xyotax
vincristine
docetaxel
vinblastine
  Intercalate DNA and inhibit RNA sysnthesis doxorubicin
dactinomycin
daunorubicin
thalidomide
  Function as a nucleoside analog gemcitabine
  Damage DNA and prevent repair bleomycin
  Inhibit DNA synthesis and RNA function cytarabine
  Inhibit dTMP synthesis 5-fluorouracil capecitabine
  Inhibit purine ring biosynthesis and dTMP
  synthesis
methotrexate
  Inhibit ribonucleotide reductase hydroxyurea
  Inhibit purine ring biosynthesis and nucleotide
  interconvesion
6-mercaptopurine 6-thioguanine
  Inhibit pyrimidine biosynthesis PALA* azaribine
  Inhibit topoisomerase I (TOPO I) irinotecan
camptothecin
lamellarin D
topotecan
karenitecin
  Inhibit topoisomerase II (TOPO II) etoposide
doxorubicin
teniposide
*

N-phosphonacetyl-L-aspartate (PALA).

TR in PHASE III PROTOCOLS

See below for highlights of some of the retrospective and prospective TR conducted in GOG phase III ovarian, primary peritoneal and/or Fallopian tube protocols that have completed accrual.

GOG 111 Protocol

GOG 111 was a Cancer Therapy Evaluation Program (CTEP)-sponsored, randomized phase III protocol by McGuire and colleagues that showed improvements in response rate (p=0.01), progression-free survival (PFS, p<0.001) and overall survival (OS; p<0.001) following intravenous paclitaxel and cisplatin compared with intravenous cyclophosphamide and cisplatin in women with previously-untreated, histologically-confirmed, suboptimal stage III EOC who underwent surgical staging and had >1 cm residual disease or stage IV EOC (Table 3) [1]. Birrer and colleagues initiated a series of retrospective studies to evaluate the prognostic relevance of a panel of markers including cell cycle regulators [2], the p53 tumor suppressor gene [3] and several proto-oncogenes including ErbB2Her2 [4] and cMYC [5] in archival formalinfixed and paraffin-embedded (FFPE) primary tumor specimens from women with advanced stage who participated in the GOG 111 protocol.

Table 3.

TR in Phase III Ovarian, Peritoneal and/or Tubal Protocols Closed to Patient Enrollment

Protocol ID Patient Population Treatments Translational Research (TR)
GOG 111 Women with previously-
untreated, histologically-
confirmed, suboptimal
stage III epithelial ovarian
cancer (EOC) who
underwent surgical staging
and had > 1 cm residual
disease or stage IV EOC
[1].
Randomized to 750 mg/m2
intravenous cyclophosphamide and
75 mg/m2 intravenous cisplatin (1
mg/minute) every 3 weeks for 6
cycles versus 135 mg/m2
intravenous paclitaxel as a
continuous 24 hour infusion on day
1 and 75 mg/m2 intravenous
cisplatin (1 mg/minute) on day 2
every 3 weeks for a 6 cycles [1].
  • Immunohistochemical (IHC) expression of cyclin E and fluorescence in situ hybridization (FISH) for cyclin E [2].

  • IHC expression of p27 {Farley et al., in progress}.

  • IHC expression of cyclin D1 and p57 {Hurteau et al., in progress}.

  • IHC expression of p53 [3].

  • FISH for Her2 and centromere of chromosome 17 (CEP17) [4].

  • FISH for c-MYC and CEP8 [5].

GOG 114 Women with previously-
untreated, histologically-
confirmed, optimally-
resected, stage III EOC
who underwent surgical
staging and had <1 cm
residual disease [6].
Randomized to a 24-hour
continuous intravenous infusion of
135 mg/m2 paclitaxel on day 1
followed by an intravenous infusion
of 75 mg/m2 cisplatin (1 mg/min) on
day 2 every 3 weeks for 6 cycles
versus intravenous carboplatin
(AUC 9.0) every 28 days for 2 cycles followed by a 24-hour
continuous intravenous infusion of
135 mg/m2 paclitaxel on day 1 and
an intraperitoneal infusion of 100
mg/m2 cisplatin on day 2 every 3
weeks for 6 cycles [6].
  • Sequencing for p53 mutations and IHC of p53 [8].

  • Relative immunoblot expression of maspin to beta-actin [9].

  • Methylation-specific PCR (MS-PCR) for the maspin promoter [9]; {Secord et al., in progress}.

  • IHC expression of maspin {Secord et al., in proqress}.

  • Relative immunoblot expression ofthrombospondin-1, bFGF, VEGF-A andVEGFR-1 to beta-actin [11].

  • Methylation-specific PCR (MS-PCR) for the thrmobospondin-1 promoter [9]; {Secord et al., in progress}.

  • Microvessel density (MVD) hotspots for CD31 and CD105 [12].

  • Relative immunoblot expression of Cyclin D1, cyclin E, cdk4, ki67, p16, Rb, p27 and p14, IHC expression of p16 and Rb, sequencing for p16 mutations and homozygous deletions [13]; {Havrilesky et al., in progress}.

  • Relative immunoblot expression of ΔNp63α to beta-actin [15].

  • Relative immunoblot expression of XRCC3 to beta-actin {Panasci et al., in progress}.

GOG 132 Women with previously-
untreated, histologically-
confirmed, suboptimally-
resected stage III and
stage IV EOC who
underwent surgical staging
and had >1 cm
residual
disease [7]
Randomized to 100 mg/m2
intravenous cisplatin (1 mg/minute)
every 3 weeks for 6 cycles versus
200 mg/m2 intravenous paclitaxel as
a continuous 24 hour infusion every
3 weeks for 6 cycles versus 135
mg/m2 intravenous paclitaxel as a
continuous 24 hour infusion followed
by 100 mg/m2 intravenous cisplatin
every 3 weeks for 6 cycles [7].
GOG 157 Women with previously
untreated, histologically-
confirmed, completely-
resected stage IA grade 3
(or clear cell tumors),
stage IB grade 3 (or clear
cell tumors), stage IC or
stage II EOC who
underwent optimal surgical
staging [17].
Randomized to a 3-hour intravenous
infusion of 175 mg/m2 paclitaxel and
a 30-minute intravenous infusion of
carboplatin (AUC7.5) every 3 weeks
for 3 versus 6 cycles [17].
  • IHC expression of p53 [3].

  • IHC expression of p27 [18]; {Farley et al., in progress}.

  • IHC expression of cyclin E [18]; {Farley et al., in progress}.

  • IHC expression of thombospondin-1, angiopoietin-1, SPARC, VEGFR-2 (Flk-1), TIE-2 and VEGF-A [19]; {Garg et al., in progress}

GOG 158 Women with previously-
untreated, histologically-
confirmed, optimal-
resected stage III EOC
who underwent adequate
surgical staging and had
<10 cm residual disease
[20].
Randomized to a 24-hour
intravenous infusion of 135 mg/m2
paclitaxel on day 1 and 75 mg/m2
intravenous cisplatin (1 mg/min) on
day 2 every 3 weeks for 6 cycles
versus a 3-hour intravenous
infusion of 175 mg/m2 paclitaxel on
day 1 and intravenous carboplatin
(AUC 7.5 mg/ml/min) on day 1
every 3 weeks for 6 cycles 20].
  • Platinum-DNA adduct level by atomic absorption spectroscopy and transcript expression of the excision repair cross complementation group 1 (ERCC1) gene by reverse transcription-polymerase chain reaction (RT-PCR) [21].

GOG 175 Women with previously-
untreated, histologically-
confirmed, completely-
resected stage IA grade 3
(or clear cell), stage IB
grade 3 (or clear cell),
stage IC or stage II EOC
who underwent optimal
surgical staging
Randomized to a 3-hour intravenous
infusion of 175 mg/m2 paclitaxel and
a 30-minute intravenous infusion of
carboplatin (AUC 6) every 3 weeks
for 3 cycles followed by a 1-hour
intravenous infusion of 40 mg/m2
every week for 24 weeks versus
observation for 24 weeks {Mannel et al.,
in progress}.
  • IHC expression of angiogenic markers and concentration of angiogenic markers in serum, plasma and urine {Kohn et al., in progress}.

  • Test of the virtual tissue banking mechnaism by examining IHC expression of Fanconi anemia complementation group (FANC), RAD51 and p53 families, and performing array-based comparative genomic hybridization (aCGH) analysis [22,23]; {Pejovic et al., in progress}.

GOG 172 Women with previously-
untreated, histologically-
confirmed, optimally-
resected, stage III EOC
who underwent adequate
surgical staging and had
<1 cm residual disease
[24].
Randomized to a 24-hour
continuous intravenous infusion of
135 mg/m2 paclitaxel on day 1
followed by 75 mg/m2 intravenous
cisplatin on day 2 every 3 weeks for
6 cycles versus a 24-hour
continuous intravenous infusion of
135 mg/m2 paclitaxel on day 1
followed by 100 mg/m2
intraperitoneal cisplatin on day 2
and 60 mg/m2 intraperitoneal
paclitaxel on day 8 every 3 weeks
for 6 cycles [24].
  • Sequencing for mutations and alterations in BRCA1 [26]; {Lesnock et al., in progress}.

  • MS-PCR for the BRCA1 promoter and transcript expression of BRCA1 by RT-PCR [27].

  • IHC expression of BRCA1 [28], {Lesnock et al., in progress}.

  • CHEK2 (CHK2) gene analysis by denaturing high-performance liquid chromatography, sequence analysis and single nucleotide polymorphism (SNP) genotyping by pyrosequencing [29].

  • SNP genotyping of codon 118 and C8092A in ERCC1 by pyrosequencing [30, 31] {Krivak et al., in progress}.

  • IHC expression of ERCC1 {Rubatt et al., in proqress}.

  • SNP genotyping of BRCA1, BRCA2, ABCB1, ABCC2, ABCG2, XRCC1 and GSTρ1 using the Sequonom iPLEXTMGOLD Assay and MALDI-TOF platform [32,33] {Tian et al., in proqress}.

  • Genome wide SNP association analysis [Birreret al., in progress; Moore et al., in progress].

GOG 182 Women with previously-
untreated, histologically-
confirmed stage III or
stage IV EOC or PPC with
either optimally-resected
disease (≤1 cm residual
disease) or suboptimally-
resected disease (>1 cm
residual disease) following
initial surgery [25].
Randomized to 3-hour intravenous
infusion of 175 mg/m2 paclitaxel on
day 1 followed by intravenous
carboplatin (AUC 6) on day 1 every
3 weeks for 8 cycles versus two
triplets and two sequential doublets
[24].*
GOG 198 Women with women with
histologically-confirimed
FIGO stage III or IV
epithelial ovarian,
Fallopian tube or primary
peritoneal cancer who
were clinically and
radiologically without
evidence of disease but
experienced biochemical recurrence as defined as a
rising CA 125 that rose to
exceed twice the upper-limit of normal limits.
Randomized to thalidomide 200 mg
oral daily dose with weekly
escalation of 100 mg to a maximum
400 mg versus tamoxifen 20 mg
oral twice daily for up to 12-months
{Hurteau et al., in progress}.
  • Enzyme-linked immunosorbent assay (ELISA) for VEGF-A in pre-cycle 1 and off-treatment serum {Hurteau et al., in progress}.

  • ELISA for VEGF-A in up to 7 serial specimens alone and in combination with CA125 {Benbrook et al., in progress}.

  • BioRad multiplex luminex assays for angiogenic markers and cytokines in serial serum specimens {Benbrook et al., in progress}.

GOG 218 Women with previously-
untreated epithelial
ovarian, primary peritoneal
or Fallopian tube cancer
who underwent adequate
surgical staging and
cytoreduction with FIGO
stage III disease with any
gross or palpable residual
disease or FIGO stage IV disease.
Randomized to a 3-hour intravenous
infusion of 175 mg/m2 paclitaxel and
intravenous carboplatin (AUC 6)
over 30 minutes on day 1 every 3
weeks for 6 cycles plus placebo (for
bevacizumab) on day 1 every 3
weeks from cycles 2 through 5
followed by placebo (for
bevacizumab) on day 1 from cycle 7
through 22 versus a 3-hour
intravenous infusion of 175 mg/m2
paclitaxel and intravenous
carboplatin (AUC 6) over 30 minutes
on day 1 every 3 weeks for 6 cycles
plus an intravenous infusion of 15
mg/kg bevacizumab on day 1 every
3 weeks from cycles 2 through 5
followed by placebo (for
bevacizumab) on day 1 from cycle 7
through 22 versus a 3-hour
intravenous infusion of 175 mg/m2
paclitaxel and intravenous
carboplatin (AUC 6) over 30 minutes
on day 1 every 3 weeks for 6 cycles
plus an intravenous infusion of 15
mg/kg bevacizumab on day 1 every
3 weeks from cycles 2 through 5
followed by an intravenous infusion
of 15 mg/kg bevacizumab on day 1
from cycle 7 through 22 {Burger et al., in progress}.
  • Genomic profiles associated with platinum-resistance, PFS and OS {Michael Birrer}.

  • Angiogenic markers in tumor and serum with potential prognostic relevance based on GOG 170D{John Fruehauf}.

  • SNPs in WNK1, GRK4 and KLKB1 associated with bevacizumab-induced hypertension {Doug Levine}.

  • Polymorphisms in codon 118 and C8092A in ERCC1 based on GOG 182 {Tom Krivak}.

  • Cell-free DNA in plasma {Anil Sood}.

  • EGFR/Her/ErbB family and ligands including soluble EGFR {Nita Maihle, Nicole Urban, Meenakshi Singh, Andre Baron}.

  • SNPs with potential predictive and prognostic clinical value {Tom Krivak, Kathleen Moore, Michael Birrer}.

*

The experimental regimens included a 3-hour intravenous infusion of 175 mg/m2 paclitaxel on day 1, intravenous 800 mg/m2/day gemcitabine on day 1 and day 8 and intravenous carboplatin (AUC 5) on day 1 every 3 weeks for 8 cycles versus a 3-hour intravenous infusion of 175 mg/m2 paclitaxel on day 1, 30 mg/m2 intravenous methoxylpolyethylene glycosylated (polyethylene glycol [PEG])-liposomal doxorubicin every other day 1 and intravenous carboplatin (AUC 5) on day 1 every 3 weeks for 8 cycles versus 1.25 mg/m2/day intravenous topotecan on day 1 and day 3 and intravenous carboplatin (AUC 5) on day 3 every 3 weeks for 4 cycles followed by a 3-hour intravenous infusion of 175 mg/m2 paclitaxel on day 1 and intravenous carboplatin (AUC 6) on day 1 every 3 weeks for 4 cycles versus 1000 mg/m2/day intravenous gemcitabine on day 1 and day 8 followed by intravenous carboplatin (AUC 6) on day 8 every 3 weeks for 4 cycles followed by a 3-hour intravenous infusion of 175 mg/m2 paclitaxel on day 1 and intravenous carboplatin (AUC 6) on day 1 every 3 weeks for 4 cycles [25].

High cyclin E protein expression, defined as >40% cyclin E positive tumor cells, was associated with a shorter median survival (29 versus 35 months) and an increased risk of death (hazard ratio [HR]=1.4, 95% confidence interval [CI]=1.0–2.1, p=0.05) [2]. This association was most notable in women with stage III disease (HR=1.7, 95% CI-1.1–2.6, p=0.03), serous histology (HR=1.8, 95% CI=1.2–2.8, p=0.01) and non-measurable disease (HR=2.4, 95% CI=1.4–4.3, p<0.01) and those randomly allocated to paclitaxel and cisplatin (HR=1.8, 95% CI=1.1–2.9) [2]. Amplification of cyclin E, detected by fluorescence in situ hybridization (FISH), was shown to be associated with high verse low cyclin E expression (p<0.006) [2]. Investigations are currently underway to evaluate the clinical utility of tumor expression of p27, cyclin D1 and p57 in the GOG 111 cohort (Table 3). Overexpression of p53, defined as ≥10% tumor cells exhibiting nuclear staining using the N-terminal DO-7 antibody (Figure 1C), was associated with GOG performance status (p=0.018) and grade (p=0.003), but not with PFS or OS [3]. ErbB2Her2 amplification, defined by FISH as >2 or >4 copies of ErbB2Her2/chromosome 17, was a rare event in EOC and was not associated with clinical characteristics, tumor characteristics or any measure of outcome including PFS or OS [4]. In addition, cMYC amplification, defined by FISH as ≥1.5 or ≥2 copies cMYC/chromosome 8, was not associated with clinical characteristics, tumor characteristics, PFS or OS [5]. Polysomy 8 was observed in 22 patients without cMYC amplification and 3 with cMYC amplification, and was associated with age and measurable disease status, but not other clinical covariates or outcomes [5].

GOG 114/132 Protocols

Berchuck and colleagues at Duke University Medical Center initiated a series of retrospective studies to evaluate the prognostic relevance of a panel of tumor suppressors, angiogenic markers, cell cycle regulators, transcriptional regulators and DNA repair proteins in frozen and archival FFPE primary tumor specimens available from women with advanced stage EOC who participated in the GOG 114 or the GOG 132 protocol. GOG 114 was a CTEP-sponsored, intergroup, randomized phase III trial with the Southwestern Oncology Group (Protocol 9227) and the Eastern Cooperative Oncology Group (Protocol GO114) by Markman and colleagues which showed improvements in PFS (p=0.01) and OS (p=0.05) with high-dose intravenous carboplatin followed by intravenous paclitaxel and intraperitoneal cisplatin compared with intravenous paclitaxel and cisplatin in women with previously-untreated, histologically-confirmed, optimally-resected, stage III EOC who underwent surgical staging and had <1 cm residual disease (Table 3) [6]. GOG 132 was a CTEP-sponsored, randomized phase III trial by Muggia and colleagues which demonstrated that inferior response rates (p<0.001) and PFS (p<0.001) but similar OS were observed with paclitaxel monotherapy compared with either cisplatin monotherapy or the paclitaxel and cisplatin combination in women with previously-untreated, histologically-confirmed, suboptimally-resected stage III and stage IV EOC who underwent surgical staging and had >1 cm residual disease (Table 3) [7].

A mutation in exons 2 to 11 of the multifunctional tumor suppressor, p53, was associated with non-mucinous or clear cell histologies (p=0.018) and a short-term reduction in the risk of disease progression (HR=0.4, 95% CI=0.2–0.8, p=0.014) and death (HR=0.3, 95% CI=0.1–0.8, p=0.014) [8]. These striking risk reductions were time-dependent and eventually disappeared around three years following initiation of primary treatment [8]. Overexpression of p53, defined as tumors with any detectable p53 immunostaining using the N-terminal DO-1 antibody (Figure 1C), was observed in 55 patients (100%) with only missense mutation(s), seven patients (32%) with truncation mutations, and eight patients (40%) lacking a mutation in exons 2 to 11, and was associated with tumor grade (p0.018) but was not associated with PFS or OS [8]. Maspin, another tumor suppressor, was not detected by immunoblot analysis in 19 (28%) of the frozen primary tumors tested, and non-detectable maspin was associated with suboptimally-debulked disease (p=0.034) and an increased risk of disease progression (HR=1.89, 95% CI=1.04–3.45, p=0.038) and death (HR=1.99, 95% CI=1.07–3.69, p=0.030) [9]. Follow up studies are underway to determine if loss of maspin expression was associated, at least in part, to methylation-induced epigenic silencing [10]. Associations were observed between categorized immunoblot expression of the pro-angiogenic factor, VEGF-A, and p53 overexpression (p=0.022), VEGFR-1 and either race (p=0.027) or histologic subtype (p=0.007), and thrombospondin-1 (an angiogenic inhibitor and promoter) and either PFS (HR=2.19, 95% CI=1.29–3.71, p=0.004) or OS (HR=1.93, 95% CI=1.12–3.32, p=0.018) [11]. High CD105 microvessel density (CD105-MVD), defined as ≥19.25 CD105 positive vessels per high density field, was associated with increased risk of disease progression (HR=1.873, 95% CI=1.102–3.184, p=0.020) but not death, whereas CD31-MVD, defined as ≥24.25 CD31-positive vessels per high power field, was not associated with PFS or OS [12]. In the GOG 114/132 cohort, none of the cancers exhibited homozygous deletions in p16, but loss of immuno-expression of p16 was associated with wild-type versus mutant p53 (p=0.03) and Rb expression (p<0.001) [13]. Investigations are currently underway to determine the optimal combination of G1 stimulators (cyclin D1, cyclin E, cdk4, Ki67) and inhibitors (p16, pRb, p27, p14) with clinical factors that predicts PFS and OS in the GOG 114/132 cohort [14]. Relative immunoblot expression of the p63 isoform lacking the transactivation domain, ΔNp63α (a homolog of p53), was associated with debulking status (p=0.023), relative expression of VEGF-A (p=0.045), and an increased risk of disease progression (HR=1.483; 95% CI=1.060–2.076; p=0.021) but not with p53 status or survival [15]. Panasci and colleagues are currently examining the prognostic relevance of x-ray repair cross-complementing protein group 3 (XRCC3), a member of RecA / RAD51-related protein family that interacts with RAD51C and is involved in DNA repair and homologous recombination to maintain chromosome stability, in the GOG 114/132 cohort.

GOG 148 Protocol

GOG 148 (Table 3) was a CTEP-sponsored, serum marker protocol by Burger and colleagues which demonstrated that among women who participated in a randomized phase III protocol for early stage EOC (GOG 95 or 157) or advanced stage EOC (GOG 111, 114, 132, 152 or 162) and had either low or high CA 125 levels, those with high sTNFR-I and low sTNFR-II levels had the lowest risk, patients with low sTNFR-I and sTNFR-II or high sTNFR-I and sTNFR-II levels had an intermediate risk, and patients with low sTNFR-I levels and high sTNFR-II levels had the highest risk of disease progression [16]. The prognostic value of serial assessment of these soluble death receptors in women with low and high CA125 is currently under investigation.

GOG 157 Protocol

GOG 157 was a CTEP-sponsored, randomized phase III trial by Bell and colleagues which demonstrated that a 6 versus 3 cycle regimen of paclitaxel and carboplatin resulted in significantly more frequent grade 3 or 4 neurotoxicity and anemia, and statistically similar PFS and OS in women with previously untreated, histologically-confirmed, completely-resected stage IA grade 3 (or clear cell tumors), stage IB grade 3 (or clear cell tumors), stage IC or stage II EOC who underwent optimal surgical staging (Table 3) [17]. Retrospective studies were also initiated to evaluate the prognostic relevance of p53 [3], cell cycle regulators [18] and angiogenic markers [19] in archival FFPE primary tumors from women who participated in the GOG 157 protocol. Overexpression of p53, defined as ≥10% tumor cells exhibiting nuclear staining using the DO-7 antibody (Figure 1C), was observed in 51% (73/143) of the GOG 157 early stage EOCs and was associated with worse PFS (logrank test: p=0.013), a 2-fold higher risk of disease progression (95% CI=1.15–3.63; p=0.015), and a similar risk of death, but was not a statistically significant independent prognostic factor for PFS (HR=1.81, 95% CI=0.99–3.30, p=0.052) [3]. Investigations are underway to examine the prognostic relevance of the IHC expression of cyclin E and p27 [18] as well as the angiogenic markers: thombospondin-1, angiopoietin-1, SPARC, VEGFR-2 (Flk-1), TIE-2 and VEGF-A [19] in the GOG 157 cohort.

GOG 158 Protocol

GOG 158 was a CTEP-sponsored, randomized phase III trial by Ozol and colleagues which demonstrated that the experimental combination of a 3-hour infusion of paclitaxel followed by carboplatin was less toxic, easier to administer and not inferior in terms of PFS and OS to the control regimen with a 24-hour infusion of paclitaxel followed by cisplatin in women with previously-untreated, histologically-confirmed, optimal-resected stage III EOC who underwent adequate surgical staging and had <1 cm residual disease (Table 3) [20]. The presence of detectable versus undetectable platinum DNA adducts was associated with longer median OS (60.3 versus 36.3 months; p=0.029) and a reduced risk of death (HR=0.607, 95% CI=0.385–0.958, p=0.032) for women with detectable versus undetectable adducts, but was not associated with PFS or an independent prognostic factor for OS [21]. ERCC1 mRNA expression, categorized as positive versus negative, in post-treatment peripheral blood leukocytes was not associated with either PFS or OS [21].

GOG 175 Protocol

GOG 175 was a CTEP-sponsored, intergroup randomized phase III trial with the Southwest Oncology Group by Mannel and colleagues that will compare PFS, OS and the frequency of adverse events in women with previously-untreated, histologically-confirmed, completely-resected stage IA grade 3 (or clear cell), stage IB grade 3 (or clear cell), stage IC or stage II EOC who were treated with intravenous paclitaxel and carboplatin every 3 weeks for 3 cycles followed by a 1-hour intravenous infusion of 40 mg/m2 every week for 24 weeks versus observation for 24 weeks (Table 3). Archival tumor specimens recovered from the GOG 175 virtual tissue bank underwent array comparative genome hybridization (aCGH) analysis to compare copy number aberrations (CNAs) in DNA repair genes including the Fanconi anemia complementation group (FANC) and RAD51 families with the rest of the genome [22,23].

GOG 172/182 Protocols

A series of TR studies are leveraging specimens and resources from the GOG 172 and 182 protocols. GOG 172 was a CTEP-sponsored, randomized phase III trial by Armstrong and colleagues which demonstrated that the intraperitoneal versus the intravenous cisplatin and paclitaxel regimen resulted in improvements in PFS (p=0.05) and OS (p=0.03) with 5.5 and 15.9 month longer median PFS and OS, respectively, worse adverse effects (p≤0.001) and quality of life before cycle 4 and 6-weeks after treatment completion, but not 1-year after treatment completion in women with previously-untreated, histologically-confirmed, optimally-resected, stage III EOC who underwent adequate surgical staging and had <1 cm residual disease (Table 3) [24]. GOG 182 was a five arm, CTEP-sponsored, international intergroup phase III randomized trial with Australia and New Zealand GOG, Medical Research Council United Kingdom; Istituto Mario Negri, Southwest Oncology Group, Eastern Cooperative Oncology Group, North Central Cancer Treatment Group, Cancer and Leukemia Group B, National Surgical Adjuvant Breast and Bowel Project and Radiation Oncology Group by Bookman and colleagues which demonstrated that addition of a third cytotoxic agent in a triplet or sequential doublet regimen following optimal or suboptimal cytoreductive surgery for the treatment of advanced stage EOC or PPC provided no added benefit to PFS or OS compared with standard paclitaxel and carboplatin in women with previously-untreated, histologically-confirmed stage III or stage IV EOC or PPC with either optimally-resected disease (≤1 cm residual disease) or suboptimally-resected disease (>1 cm residual disease) following initial surgery (Table 3) [25].

Thus far, mutations in BRCA1 were identified in DNA extracted from a buffy coat specimen from 16 (5%) of GOG 172 patients [26]. A thorough evaluation of the type and distribution of mutations and common variations observed in BRCA1 and associations with clinical outcome in the GOG 172 cohort are currently underway. BRCA1 promoter methylation was observed in specific CpG sites in sporadic EOC including women who participated in GOG 172 and transcript expression of BRCA1 by RT-PCR was significantly lower in women with a methylated compared with an unmethylated BRCA1 promoter [27]. Low IHC expression of BRCA1, defined as <10% positive tumor cells, was associated with advanced stage (p<0.001), serous histology (p<0.001), better PFS (p=0.03) and OS (p=0.006), and a reduced risk of disease progression (HR=0.64, 95% CI=0.42–0.96) and death (HR=0.51, 95% CI=0.32–0.83) [28]. Results of additional IHC studies of BRCA1 by route of administration in GOG 172 cases will be presented at the 2010 Society of Gynecologic Oncologist (SGO) Meeting. Denaturing high-performance liquid chromatography, sequence analysis, and single nucleotide polymorphism genotyping by pyrosequencing for the CHEK2 gene demonstrated that variations in CHEK2 do not appear to make a significant contribution to the pathogenesis of sporadic EOC in the United States [29]. In the GOG 172 cohort, the codon 118 polymorphism in ERCC1 was not significantly associated with disease progression or death whereas the C/A or A/A versus C/C genotypes in C8092A in ERCC1 were associated with 6- and 17-month shorter median PFS and OS, respectively, and an increased risk of disease progression (HR=1.44, 95% CI=1.06–1.94, p=0.018) and death (HR=1.50, 95% CI=1.07–2.09, p=0.018) [30]. Subset analysis stratified by treatment regimen demonstrated a distinct PFS and OS advantage for women with the C/C compared with either the C/A or A/A genotypes in C8092A in ERCC1 in women randomly allocated to the IP treatment arm [30]. The associations between codon 118 and C8092A polymorphisms in ERCC1 and PFS and OS in the GOG 182 cohort are under active investigation [31]. Results of an IHC study of ERCC1 in GOG 172 will be presented at the 2010 SGO Meeting. Additional studies are also underway to evaluate the relationship between common polymorphisms in DNA repair genes (BRCA1 and BRCA2 [32] as well as XRCC1), efflux pumps (ABCB1, ABCC2 and ABCG2 [33]) and detoxicification enzyme (GSTρ1) and measures of clinical outcome including PFS, OS and common severe adverse effects, and to perform genome-wide association studies in the GOG 172 and 182 cohorts.

GOG 198 Protocol

GOG 198 was a CTEP-sponsored, randomized phase III trial by Hurteau and colleagues that evaluated oral daily thalidomide versus tamoxifen and the prognostic relevance of serum VEGF in women with biochemical-recurrent epithelial ovarian, fallopian tube or primary peritoneal cancer (Table 3). Investigations are also underway to evaluate the serial changes in VEGF-A during treatment and other angiogenic markers and cytokines in this setting.

GOG 218 Protocol

GOG 218, sponsored by CTEP and Genetech, is a three-arm, randomized, doubleblinded, placebo-controlled phase III trial by Burger and colleagues that will evaluate paclitaxel, carboplatin and placebo plus placebo maintenance versus paclitaxel, carboplatin and bevacizumab plus placebo maintenance versus paclitaxel, carboplatin and bevacizumab plus bevacizumab in women with previously-untreated epithelial ovarian, primary peritoneal or fallopian tube cancer who underwent adequate surgical staging and cytoreduction with FIGO stage III disease with any gross or palpable residual disease or FIGO stage IV disease (Table 3). A series of TR studies are leveraging specimens and resources including those designed to validate published genomic profiles, angiogenic markers, single nucleotide polymorphisms (SNPs), cell-free DNA, EGFR family members/ligands, markers of VEGF-targeted therapy as well as new markers and profiles including those identified by The Genome Atlas Project in ovarian cancer. New discovery and validation efforts are also built into GOG 218 including a number of genomic profiling, SNP and genome-wide association studies.

TR in DEVELOPMENTAL THERAPEUTICS PROTOCOLS

The GOG has conducted TR in a series of phase II ovarian, primary peritoneal and/or Fallopian tube protocols (sponsor) including the evaluation of capecitabine in GOG 146L (CTEP/Roche Laboratories) [34], bortezomib in GOG 146N (CTEP/Millennium Pharmaceuticals, Inc.) [35], cetuximab and carboplatin in 146P (Bristol-Myers Squibb/Imclone) [36], trastuzumab in GOG 160 (CTEP/Genetech) [37], gefitinib in 170C (CTEP) [38], bevacizumab in GOG 170D (CTEP) [39,40], imatinib in GOG 170E (CTEP/Novartis) [41], sorafenib in GOG 170F (CTEP), lapatinib in GOG 170G (CTEP), temsirolimus in GOG 170I (CTEP), enzastaurin in GOG 170J (Eli Lilly), AMG 706 in GOG 170L (Amgen), dasatinib in GOG 170M (Bristol-Myers Squibb), and A6 in GOG 170N (Angstrom). Results of 170I and 170J will be presented at the 2010 SGO Meeting. Other phase II protocols with TR are in various stages of development and design including the evaluation of AMG 102 in GOG 170P (Amgen), EGEN-001 in DTM0835 (Expression Genetics), TRC105 in DTM0917 (Tracon Pharmaceuticals), MK-2206 in DTM0926 (Merck) and MK-4827 in DTM-0929 (Merck). Table 1 provides details regarding the mechanism(s) of action of the molecular targeting agents.

ADDITIONAL TR STUDIES

GOG 136 Protocol

GOG 136, is a CTEP-sponsored, specimen banking protocol by Cibull and colleagues for women undergoing a surgery for a gynecologic malignancy or a prophylactic oophorectomy which has supported a number of note worthy studies including those reported by Zorn et al. [42,43] and Beck et al. [4446].

GOG 143/144 Protocols

The CTEP-sponsored GOG 143 protocol demonstrated that in an unselected, clinic-based series of ovarian cancer cases, 12 patients exhibited protein truncation mutations in BRCA1 and another 12 displayed BRCA1 mutations of unknown significance [47]. GOG 144 was a CTEP-sponsored protocol for women with familial ovarian cancer that screened 26 women for mutations in BRCA1/BRCA2 and detected 12 deleterious alterations; 8 in BRCA1 and 4 in BRCA2 [48]. Mutations in BRCA1 or BRCA2 were present in about 50% of ovarian cancer patients with at least one first-degree relative with disease, and in 70% of patients with two or more relatives with ovarian/breast cancer (p=0.0002) [48].

GOG 199 Protocol

The GOG 199 protocol is a prospective, international, two-cohort, non-randomized study by Greene and colleagues, in the Clinical Genetics Branch an NCI Intramural Research Program, the GOG, and the Cancer Genetics Network, in women at genetic risk of ovarian cancer, who undergo risk-reducing salpingo-oophorectomy (RRSO) or screening [49]. A series of TR studies leverage specimens and resources from the GOG 199 protocol including GOG 246CIMBA-4 which demonstrated that the minor allele of the rs3817198 SNP in LSP1 was associated with increased breast cancer risk only for BRCA2 mutation carriers (HR=1.16, 95%CI=1.07–1.25, p-trend=2.8×10−4) whereas the rs13387042 SNP at 2q35, but not the rs13281615 SNP at 8q24, was associated with breast cancer risk for BRCA1 mutation carriers (HR=1.14, 95% CI=1.04–1.25, p=0.005) and BRCA2 mutation carriers (HR=1.18, 95%CI=1.04–1.33, p=.0079) [50]. In addition, GOG 8008CIMBA-5 will evaluate the association between SNPs in rs16942 in BRCA1, rs2237060 in RAD50, SNP3 and rs2241193 in IGFBP5 and breast cancer risk in BRCA1/BRCA2 mutation carriers. GOG 8009 and 8010 represent genome-wide association studies examining modifiers of breast cancer risk in BRAC1 and BRCA2 mutation carriers, respectfully. Additional studies are also under development including CIMBA-6 and CIMBA-7.

FUTURE DIRECTIONS

Release of data from the TCGA project in ovarian cancer is expected to solidify our view of ovarian cancer by defining a comprehensive catalog of the genomic and epigenetic changes in EOC. Translation of these findings into clinical trials and practice will require coordinated efforts that leverage resources, expertise and funding for retrospective and prospective validation studies in well-annotated specimens from independent EOC patients. The GOG offers a variety of mechanisms and unique sets of specimens and resources to translate the TCGA findings into marker-driven phase II and ultimately phase III clinical trials that advance molecular oncology and personalized medicine for women with EOC and establish new standards of care with an arsenal of validated markers and profiles with diagnostic, prognostic and/or predictive value.

ACKNOWLEDGEMENTS

The GOG patients, leadership, committee chairs, study chairs, laboratory and scientific collaborators, statisticians, translational research scientists, clinical data coordinators, programmers, members and sponsors as well as staff in the GOG Statistical and Data Center, Administrative Office, Finance/Development Office, Tissue Bank and institutions who participated in the GOG studies reported in this review are recognized for their efforts and contributions to these clinical and/or translational research studies.

This review was supported by National Cancer Institute (NCI) grants to the Gynecologic Oncology Group (GOG) Administrative Office (CA 27469) and the GOG Statistical and Data Center (CA 37517).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

PRECIS

Advances in molecular oncology and personalized medicine for women with epithelial ovarian cancer are being realized through the evaluation of markers, pathways and novel therapies.

CONFLICT OF INTEREST STATEMENT

There are no conflicts of interest except that Dr. Darcy is an employee of the Gynecologic Oncology Group and the organization receives funding from various organizations including the Cancer Therapy Evaluation Program, the National Cancer Institute at the National Institute of Health and Industry collaborators who participated in and/or sponsored the protocols reported in this review.

REFERENCES

  • 1.McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look KY, et al. Cyclophosphamide and cisplatin compared with paclitaxel and cisplatin in patients with stage III and stage IV epithelial ovarian cancer. N Engl J Med. 1996;334(1):1–6. doi: 10.1056/NEJM199601043340101. [DOI] [PubMed] [Google Scholar]
  • 2.Farley J, Smith LM, Darcy KM, Sobel E, O'Connor D, Henderson B, Morrison LE, Birrer MJ. Cyclin E expression is a significant predictor of survival in advanced, suboptimally debulked ovarian epithelial cancers: a Gynecologic Oncology Group study. Cancer Res. 2003;63:1235–1241. [PubMed] [Google Scholar]
  • 3.Darcy KM, Brady WE, McBroom JW, Bell JG, Young RC, McGuire WP, et al. Associations between p53 overexpression and multiple measures of clinical outcome in high-risk, early stage and suboptimally-resected, advanced stage epithelial ovarian cancers: A Gynecologic Oncology Group study. Gynecol Oncol. 2008;111(3):487–495. doi: 10.1016/j.ygyno.2008.08.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Farley J, Fuchiuji S, Darcy KM, Tian C, Hoskins WJ, McGuire WP, et al. Associations between ERBB2 amplification and progression-free survival and overall survival in advanced stage, suboptimally-resected epithelial ovarian cancers: A Gynecologic Oncology Group study. Gynecol Oncol. 2009;113(3):341–347. doi: 10.1016/j.ygyno.2009.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Darcy KM, Brady WE, Blancato JK, Dickson RB, Hoskins WJ, McGuire WP, et al. Prognostic relevance of c-MYC gene amplification and polysomy for chromosome 8 in suboptimally-resected, advanced stage epithelial ovarian cancers: A Gynecologic Oncology Group study. Gynecol Oncol. 2009;114(3):472–479. doi: 10.1016/j.ygyno.2009.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Markman M, Bundy BN, Alberts DS, Fowler JM, Clarke-Pearson DL, Carson LF, et al. Phase III trial of standard-dose intravenous cisplatin plus paclitaxel versus moderately high-dose carboplatin followed by intravenous paclitaxel and intraperitoneal cisplatin in small-volume stage III ovarian carcinoma: an intergroup study of the Gynecologic Oncology Group, Southwestern Oncology Group, and Eastern Cooperative Oncology Group. J Clin Oncol. 2001;19(4):1001–1007. doi: 10.1200/JCO.2001.19.4.1001. [DOI] [PubMed] [Google Scholar]
  • 7.Muggia FM, Braly PS, Brady MF, Sutton G, Niemann TH, Lentz SL, Alvarez RD, Kucera PR, Small JM. Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: A Gynecologic Oncology Group Study. J Clin Oncol. 2000;18(1):106–115. doi: 10.1200/JCO.2000.18.1.106. [DOI] [PubMed] [Google Scholar]
  • 8.Havrilesky L, Darcy KM, Hamdan H, Priore RL, Leon J, Bell J, Berchuck A. Prognostic significance of p53 mutation and p53 overexpression in advanced epithelial ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol. 2003;21(20):3814–3825. doi: 10.1200/JCO.2003.11.052. [DOI] [PubMed] [Google Scholar]
  • 9.Secord AA, Lee PS, Darcy KM, Havrilesky LJ, Grace LA, Marks JR, Berchuck A. Maspin expression in epithelial ovarian cancer and associations with poor prognosis: A Gynecologic Oncology Group study. Gynecol Oncol. 2006;101(3):390–397. doi: 10.1016/j.ygyno.2006.02.014. [DOI] [PubMed] [Google Scholar]
  • 10.Secord AA, Lee PS, Jewell E, Havrilesky L, Grace L, Murphy Berchuck A, Darcy KM, Hutson A. The role of methylation in the regulation of angiogenic inhibitors in advanced epithelial ovarian cancer: a Gynecologic Oncology Group Study. Gynecol Oncol. 2006;101:S84. [Google Scholar]
  • 11.Secord AA, Darcy KM, Hutson A, Lee PS, Havrilesky LJ, Grace LA. Berchuck A. Co-expression of angiogenic markers and associations with prognosis in advanced epithelial ovarian cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2007;106(1):221–232. doi: 10.1016/j.ygyno.2007.03.021. [DOI] [PubMed] [Google Scholar]
  • 12.Rubatt JM, Darcy KM, Hutson A, Bean S, Havrilesky LJ, Grace LA, et al. Independent prognostic relevance of microvessel density in advanced epithelial ovarian cancer and associations between CD31, CD105, p53 status and angiogenic marker expression: a Gynecologic Oncology Group study. Gynecol Oncol. 2009;112(3):469–474. doi: 10.1016/j.ygyno.2008.11.030. [DOI] [PubMed] [Google Scholar]
  • 13.Havrilesky L, Alvarez A, Whitaker R, Marks J, Berchuck A. Loss of expression of the p16 tumor suppressor gene is more frequent in advanced ovarian cancers lacking p53 mutations: a Gynecologic Oncology Group study. Gynecol Oncol. 2001;80(2):283. doi: 10.1006/gyno.2001.6464. [DOI] [PubMed] [Google Scholar]
  • 14.Havrilesky LJ, Fleming ND, Miner Z, Darcy K, Hutson A, Berchuck A, et al. Correlations between cell cycle regulatory genes and relationship to ovarian cancer prognosis: a Gynecologic Oncology Group study. Gynecol Oncol. 2007;104:S54. [Google Scholar]
  • 15.Jewell EL, Darcy KM, Hutson A, Lee PS, Havrilesky LJ, Grace LA, Berchuck A, Alvarez Secord A. Association between the N-terminally truncated (ΔN) p63α (ΔNp63α) isoform and debulking status, VEGF expression and progression-free survival in previously-untreated, advanced stage epithelial ovarian cancer: A Gynecologic Oncology Group study. Gynecol Oncol. 2009;115(3):424–429. doi: 10.1016/j.ygyno.2009.07.035. [DOI] [PubMed] [Google Scholar]
  • 16.Burger RA, Darcy KM, DiSaia PJ, Monk BJ, Grosen EA, Gatanaga T, et al. Association between serum levels of soluble tumor necrosis factor receptors/CA 125 and disease progression in patients with epithelial ovarian malignancy: a Gynecologic Oncology Group study. Cancer. 2004;101(1):106–115. doi: 10.1002/cncr.20314. [DOI] [PubMed] [Google Scholar]
  • 17.Bell J, Brady M, Young R, Lage J, Walker J, Look KY, et al. Randomized phase III trial of three versus six cycles of adjuvant carboplatin and paclitaxel in early stage ovarian carcinoma: a Gynecologic Oncology Group study. Gynecol Oncol. 2006;102(3):432–439. doi: 10.1016/j.ygyno.2006.06.013. [DOI] [PubMed] [Google Scholar]
  • 18.Farley JH, Smith L, Darcy KM, Tian C, Sobel E, Birrer MJ. Alterations in cyclin E and p27 in early epithelial ovarian cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2002;84(3):499. [Google Scholar]
  • 19.Garg R, Garcia RL, Brady MF, Goff BA, Paley P. Angiogenic marker alteration in high-risk stage I or II epithelial ovarian cancer. Gynecol Oncol. 2007;104(3):S43. [Google Scholar]
  • 20.Ozol RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson, Burger RA, Mannel RS, DeGeest K, Hartenbach EM, Baergen R. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: A Gynecologic Oncology Group study. J Clin Oncol. 2003;21(17):3194–3200. doi: 10.1200/JCO.2003.02.153. [DOI] [PubMed] [Google Scholar]
  • 21.Darcy KM, Tian C, Reed E. A Gynecologic Oncology Group study of platinum-DNA adducts and excision repair cross-complementation group 1 expression in optimal, stage III epithelial ovarian cancer treated with platinum-taxane chemotherapy. Cancer Res. 2007;67(9):4474–4481. doi: 10.1158/0008-5472.CAN-06-4076. [DOI] [PubMed] [Google Scholar]
  • 22.Pejovic T, Gaile DP, Darcy KM, Liu S, Shepherd L, Rodgers WH, Kohn E, Mannel R, Birrer MJ, Nowak N. A Gynecologic Oncology Group study of frequent copy number aberrations in DNA repair genes and other genomic regions in stage I serous ovarian cancers. J Clin Oncol. 2009;27 (ASCO Abstract #e16504) [Google Scholar]
  • 23.Gaile DP, Shepherd L, Liu S, Darcy K, Brady M, Morrison C. iGenomic Viewer, a Gynecologic Oncology Group software library for the creation of highly customizable, portable, interactive, and linked visualizations of high throughput genomic data. J Clin Oncol. 2009;27 (ASCO Abstract # e16544) [Google Scholar]
  • 24.Armstrong DK, Bundy BN, Wenzel L, Huang HQ, Baergen R, Lele SB, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med. 2006;354(1):34–43. doi: 10.1056/NEJMoa052985. [DOI] [PubMed] [Google Scholar]
  • 25.Bookman M, Brady M, McGuire W, Harper P, Alberts D, Friedlander M, et al. Evaluation of new platinum-based treatment regimens in advanced-stage ovarian cancer: a phase III trial of the Gynecologic Cancer Intergroup. J Clin Oncol. 2009;27(9):1419–1425. doi: 10.1200/JCO.2008.19.1684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gallion H, Kryscio R, Clarke-Pearson D, DiSaia P, Fowler J, Mannel R, et al. The prognostic significance of germline BRCA1 mutations in epithelial ovarian cancer: a Gynecologic Oncology Group Study, Gynecol Oncol. 2003;88(2):157. [Google Scholar]
  • 27.Wilcox CB, Baysal BE, Gallion HH, Strange MA, DeLoia J. High-resolution methylation analysis of the BRCA1 promoter in ovarian tumors. Cancer Genet Cytogenet. 2005;159(2):114–122. doi: 10.1016/j.cancergencyto.2004.12.017. [DOI] [PubMed] [Google Scholar]
  • 28.Thrall M, Gallion HH, Kryscio R, Kapali M, Armstrong DK, Deloia JA. BRCA1 expression in a large series of sporadic ovarian carcinomas: a Gynecologic Oncology Group study. Int J Gynecol Cancer. 2006;16 Suppl. 1:166–171. doi: 10.1111/j.1525-1438.2006.00504.x. [DOI] [PubMed] [Google Scholar]
  • 29.Baysal BE, DeLoia JA, Willett-Brozick JE, Goodman MT, Brady MF, Modugno F, et al. Analysis of CHEK2 gene for ovarian cancer susceptibility. Gynecol Oncol. 2004;95(1):62–69. doi: 10.1016/j.ygyno.2004.07.015. [DOI] [PubMed] [Google Scholar]
  • 30.Krivak TC, Darcy KM, Tian C, Armstrong D, Baysal BE, Gallion H, Ambrosone CB, DeLoia JA. Relationship between ERCC1 polymorphisms and 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(21):3598–3606. doi: 10.1200/JCO.2008.16.1323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Krivak TC, Darcy K, Tian C, Bookman MA, Baysal BA, Gallion H, et al. Relationship between ERCC1 polymorphism, disease progression and survival in GOG0182: a Gynecologic Oncology Group phase III trial of platinum-based chemotherapy in women with advanced stage epithelial ovarian or primary peritoneal cancer. J Clin Oncol. 2008;26(15s):302s. [Google Scholar]
  • 32.Krivak TC, Tian C, DeLoia JA, Darcy KM, Armstrong D, Gallion H, et al. BRCA1/BRCA2 polymorphisms and prognosis in women with optimally-resected stage III epithelial ovarian cancer treated on GOG Protocol 172: A Gynecologic Oncology Group study. Gynecol Oncol. 2009;112(2):S9. [Google Scholar]
  • 33.Darcy KM, Tian C, Ambrosone CB, Krivak TC, Armstrong D, Bookman MA, et al. A Gynecologic Oncology Group study of associations between polymorphisms in ABC transporter genes (ABCB1, ABCC2 and ABCG2) and outcome in advanced stage epithelial ovarian cancer treated with platinum and taxane chemotherapy. J Clin Oncol. 2009;27(15S):s293. [Google Scholar]
  • 34.Garcia AA, Blessing JA, Lenz HJ, Darcy KM, Mannel RS, Miller DS, et al. Phase II clinical trial of capecitabine in ovarian carcinoma recurrent 6–12 months after completion of primary chemotherapy, with exploratory TS, DPD, and TP correlates: a Gynecologic Oncology Group study. Gynecol Oncol. 2005;96(3):810–811. doi: 10.1016/j.ygyno.2004.11.037. [DOI] [PubMed] [Google Scholar]
  • 35.Aghajanian C, Blessing JA, Darcy KM, Reid G, DeGeest K, Rubin SC, et al. A phase II evaluation of bortezomib in the treatment of recurrent platinum-sensitive ovarian or primary peritoneal cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2009;115(2):215–220. doi: 10.1016/j.ygyno.2009.07.023. [DOI] [PubMed] [Google Scholar]
  • 36.Secord AA, Blessing JA, Armstrong DK, Rodgers WH, Miner Z, Barnes MN, Lewandowski G, Mannel RS. Phase II trial of cetuximab and carboplatin in relapsed platinum-sensitive ovarian or cancer and evaluation of epidermal growth factor receptor expression: a Gynecologic Oncology Group study. Gynecol Oncol. 2008;108(3):493–499. doi: 10.1016/j.ygyno.2007.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bookman MA, Darcy KM, Clarke-Pearson D, Boothby RA, Horowitz IR. Evaluation of monoclonal humanized anti-HER2 antibody (Trastuzumab) in patients with recurrent or refractory ovarian or primary peritoneal carcinoma with overexpression of HER2: a phase II trial of the Gynecologic Oncology Group. J.Clin.Oncol. 2003;21(2):283–290. doi: 10.1200/JCO.2003.10.104. [DOI] [PubMed] [Google Scholar]
  • 38.Schilder R, Sill M, Chen X, Darcy K, DeCesare S, Lewandowski G, et al. Phase II study of gefitinib in patients with relapsed or persistent ovarian or primary peritoneal carcinoma and evaluation of epidermal growth factor receptor mutations and immunohistochemical expression: A Gynecologic Oncology Group study. Clin Cancer Res. 2005;11(15):5539–5548. doi: 10.1158/1078-0432.CCR-05-0462. [DOI] [PubMed] [Google Scholar]
  • 39.Burger RA, Sill MW, Monk BJ, Greer BE, Sorosky JI. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: a Gynecologic Oncology Group Study. J Clin Oncol. 2007;25(33):5165–5171. doi: 10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
  • 40.Han ES, Burger R, Darcy KM, Sill M, Greer BE, Sorosky JI, Fruehauf JP. Relationship between angiogenic markers and clinicopathologic factors/outcome in GOG-170D, a phase II trial of bevacizumab (Bev) in recurrent or persistent epithelial ovarian cancer (EOC) and primary peritoneal cancer (PPC) J Clin Oncol. 2008;26(15S) [Google Scholar]
  • 41.Schilder RJ, Sill MW, Lee RB, Shaw TJ, Senterman MK, Klein-Szanto AJP, et al. Phase II evaluation of imatinib methylate in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma: a Gynecologic Oncology Group study. J Clin Onco.l. 2008;26(20):3418–3425. doi: 10.1200/JCO.2007.14.3420. [DOI] [PubMed] [Google Scholar]
  • 42.Zorn KK, Jazaeri AA, Awtrey CS, Gardner GJ, Mok SC, Goyd J, et al. Choice of normal ovarian control influences determination of differentially expressed genes in ovarian cancer expression profiling studies. Clin Cancer Res. 2003;9(13):4811–4818. [PubMed] [Google Scholar]
  • 43.Zorn KK, Bonome T, Gangi L, Chandramouli GV, Awtrey CS, Gardner GJ, et al. Gene expression of serous, endometrioid, and clear cell types of ovarian and endometrial cancer. Clin Cancer Res. 2005;11(18):6422–6430. doi: 10.1158/1078-0432.CCR-05-0508. [DOI] [PubMed] [Google Scholar]
  • 44.Mo YY, Yu Y, Theodosiou E, Ee PL, Beck WT. A role for Ubc9 in tumorigenesis. Oncogene. 2007;24(16):2677–2683. doi: 10.1038/sj.onc.1208210. [DOI] [PubMed] [Google Scholar]
  • 45.He X, Ee PL, Coon J, Beck WT. Alternative splicing of the multidrug resistance protein 1/ATP binding cassette transporter subfamily gene in ovarian cancer creates functional splice variants and is associated with increased expression of the splicing factors PTB and SRp20. Clin Cancer Res. 2004;10(4):4652–4660. doi: 10.1158/1078-0432.CCR-03-0439. [DOI] [PubMed] [Google Scholar]
  • 46.He X, Pool M, Darcy KM, Lim SB, Auersperg N, Coon JS, et al. Knockdown of polypyrimidine tract-binding protein suppresses ovarian tumor cell growth and invasiveness in vitro. Oncogene. 2007;26(34):4961–4968. doi: 10.1038/sj.onc.1210307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Smith SA, Richards WE, Caito K, Hanjani P, Markman M, Degeest K, et al. BRCA1 germline mutations and polymorphisms in a clinic-based series of ovarian cancer cases: a Gynecologic Oncology Group study. Gynecol Oncol. 2001;83(3):586–592. doi: 10.1006/gyno.2001.6430. [DOI] [PubMed] [Google Scholar]
  • 48.Reedy M, Gallion H, Fowler J, Kryscio R, Smith S. Contribution of BRCA1 and BRCA2 to familial ovarian cancer: a Gynecologic Oncology Group study. Gynecol Oncol. 2002;85(2):255–259. doi: 10.1006/gyno.2002.6615. [DOI] [PubMed] [Google Scholar]
  • 49.Greene MH, Piedmonte M, Alberts D, Gail M, Hensley M, Miner Z, et al. A prospective study of risk-reducing salpingo-oophorectomy and longitudinal CA-125 screening among women at increased genetic risk of ovarian cancer: design and baseline characteristics: A Gynecologic Oncology Group study. Cancer Epidemiol Biomarkers Prev. 2008;17(3):594–604. doi: 10.1158/1055-9965.EPI-07-2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Antoniou AC, Sinilnikova O, McGuffog L, Healey S, Nevanlinna H, Simard J, et al. Common variants in 8q24, LSP1 and 2q35 and breast cancer risk for BRCA1 and BRCA2 mutation carriers. Hum Mol Genet. 2009;18(22):4442–4456. doi: 10.1093/hmg/ddp372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Darcy KM, Wohlhueter AL, Zangani D, Vaughan MM, Russell JA, Masso-Welch PA, et al. Selective changes in EGF receptor expression and function during the proliferation, functional differentiation and apoptosis of mammary epithelial cells. Euro J Cell Biol. 1999;78:511–523. doi: 10.1016/S0171-9335(99)80077-6. [DOI] [PubMed] [Google Scholar]
  • 52.Darcy KM, Zangani D, Wohlhueter AL, Huang R-Y, Vaughan MM, Russell JA, Ip MM. Changes in ErbB2 (HER-2/neu), ErbB3 and ErbB4 during the growth, differentiation and apoptosis of normal rat mammary epithelial cells. J Histochem Cytochem. 2000;48(1):63–80. doi: 10.1177/002215540004800107. [DOI] [PubMed] [Google Scholar]
  • 53. http://www.sigmaaldrich.com/life-science/cell-biology/learning-center/pathway-slides-and/signaling-pathways.html.
  • 54. http://www.sigmaaldrich.com/life-science/cell-biology/learning-center/pathway-slides-and/regulatory-cascade-of-cyclin-gene-expression.html.
  • 55. http://www.sigmaaldrich.com/life-science/cell-biology/learning-center/pathway-slides-and/g1-and-s-phases-of-the-cell-cycle.html.
  • 56.Vilmar A, Sørensen JB. Excision repair cross-complementation group 1 (ERCC1) in platinum-based treatment of non-small cell lung cancer with special emphasis on carboplatin: a review of current literature. Lung Cancer. 2009;64(2):131–139. doi: 10.1016/j.lungcan.2008.08.006. [DOI] [PubMed] [Google Scholar]
  • 57.Durant ST, Nickoloff JA. Good timing in the cell cycle for precise DNA repair by BRCA1. Cell Cycle. 2005;4(9):1216–1222. doi: 10.4161/cc.4.9.2027. [DOI] [PubMed] [Google Scholar]
  • 58.Weberpals JI, Clark-Knowles KV, Vanderhyden BC. Sporadic epithelial ovarian cancer: clinical relevance of BRCA1 inhibition in the DNA damage and repair pathway. J Clin Oncol. 2008;26(19):3259–3267. doi: 10.1200/JCO.2007.11.3902. [DOI] [PubMed] [Google Scholar]
  • 59.Bolderson E, Richard DJ, Zhou B-BS, Khanna KK. Recent advances in cancer therapy targeting proteins involved in DNA double-strand repair. Clin Cancer Res. 2009;15(20):6314–6320. doi: 10.1158/1078-0432.CCR-09-0096. [DOI] [PubMed] [Google Scholar]

RESOURCES