Hindawi Publishing Corporation
Journal of Oncology
Volume 2010, Article ID 149362, 11 pages
doi:10.1155/2010/149362
Review Article
A Current Review of Targeted Therapeutics for Ovarian Cancer
Susana M. Campos1 and Sue Ghosh2
1 Dana
Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
2 Brigham
Correspondence should be addressed to Susana M. Campos,
[email protected]
Received 9 July 2009; Accepted 28 September 2009
Academic Editor: Maurie M. Markman
Copyright © 2010 S. M. Campos and S. Ghosh. This is an open access article distributed under the Creative Commons Attribution
License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Difficult to detect, ovarian cancer typically presents at an advanced stage. Significant progress has been achieved in the treatment
of ovarian cancer with therapeutics focused on DNA replication or cell division. However, despite sensitivity to induction
chemotherapy the majority of patients will develop recurrent disease. Conventional agents for recurrent disease offer little in
terms of long-term responses. Various targeted therapeutics have been explored in the management of ovarian cancer. These
include monoclonal antibodies to epidermal growth factor receptors, small molecule tyrosine kinase inhibitors, monoclonal
antibodies directed at the vascular endothelial growth factor (bevacizumab), and the small tyrosine kinase inhibitors that target the
vascular endothelial growth factor receptor. Recently, several other agents have come forth as potential therapeutic agents in the
management of ovarian cancer. These include monoclonal antibodies to the folate receptor, triple angiokinase inhibitors, PARP
inhibitors, aurora kinase inhibitors, inhibitors of the Hedgehog pathway, folate receptor antagonists, and MTOR inhibitors.
1. Introduction
2. Angiogenesis
Various targeted therapeutics have been explored in the
management of ovarian cancer. These include monoclonal
antibodies to Her 2 neu [1, 2] and other epidermal growth
factor receptors [3] (i.e., Trastuzumab [1], Pertuzumab
[2], and EMD 7200 [3]), small molecule tyrosine kinase
inhibitors that targeted the various EGFR receptors (gefitinib
[4], erlotinib [5], CI-1033 [6]), monoclonal antibodies
directed at the vascular endothelial growth factor [7–19]
(bevacizumab), and the small tyrosine kinase inhibitors
that target the vascular endothelial growth factor receptor
[20–25]. Recently, several other agents have come forth as
potential therapeutic agents in the management of ovarian
cancer. These include monoclonal antibodies to the folate
receptor, triple angiokinase inhibitors, PARP inhibitors,
aurora kinase inhibitors, inhibitors of the Hedgehog pathway, folate receptor antagonists, and MTOR inhibitors.
This paper will explore the current data on the various
targeted approaches in ovarian cancer. Attention will be
directed at understanding the molecular mechanisms of
these agents balanced with their application to clinical
practice.
Enthusiasm for cytotoxic agents in the management of
ovarian cancer has been tempered by the emergence of
resistance. As such, a focus on alternative innovative therapeutics has emerged. One such direction is the inhibition of
angiogenesis. Angiogenesis is one of the cardinal processes
leading to invasion and metastasis of solid tumors. The
angiogenic-signaling pathway may be triggered by the release
of angiogenic ligands such as the vascular endothelial
growth factor from tumor cells. Tumor angiogenesis is well
established as essential for the growth and metastasis of solid
tumors, [26–28] This process involves the recruitment of
mature vasculature and circulating endothelial cells [29, 30]
and proangiogenic soluble mediators one of which includes
the vascular endothelial growth factor (VEGF) [31]. This
factor has several known activities [31], such as mitogenesis,
angiogenesis, endothelial survival, enhancement of vascular
permeability, and effects on hemodynamic status. In ovarian
cancer increased levels of VEGF are associated with poor
prognosis and have been confirmed in multivariate analysis
as an independent prognostic indicator of survival [28, 32–
38]. Given the poor long-term responses appreciated with
2
conventional cytotoxic agents that target VEGF have taken
center stage.
Agents targeting angiogenesis include monoclonal antibodies to the VEGF ligand [7–19], small tyrosine kinase
inhibitors that target the vascular endothelial growth factor
receptor [20–25], and soluble decoy VEGF receptors [39, 40].
The most studied agent to date has been bevacizumab, a
recombinant humanized monoclonal antibody to the VEGF
ligand.
To date several investigators [7–19] (Table 1) have
explored bevacizumab as a single agent or in combination
with chemotherapy in the management of advanced ovarian
cancer.
Several studies in both the upfront and in the recurrent
setting are underway. GOG 218 is a randomized placebo
controlled three-arm study examining the role of bevacizumab in combination with carboplatin and paclitaxel
and also as a maintenance therapy. ICON-7 is a two arm
trial comparing carboplatin and paclitaxel (six cycles) versus
carboplatin, paclitaxel, and bevacizumab (7.5 mg/kg) for six
cycles followed by 12 cycles of maintenance bevacizumab.
Campos et al. [20] is conducting a phase II trial of carboplatin/paclitaxel/bevacizumab in optimally and suboptimally
debulked patients. Patients achieving a clinical complete
response, partial response, or stable disease are subsequently
randomized to either bevacizumab for 12 months or the
combination of bevacizumab and erlotinib. Preliminary
safety results have noted an increase in hypertension but to
date no evidence of gastrointestinal perforations.
Given the recent data that has emerged on the role
on intraperitoneal chemotherapy [41–43] investigators are
exploring the role of IP chemotherapy with IV bevacizumab.
Several abstracts were highlighted at the recent American
Society of Clinical Oncology meeting. Konner et al. [44]
and McKeekin et al. [45] in independent studies reported
the feasibility of utilizing bevacizumab (IV) in conjunction
with intraperitoneal therapy. One bowel perforation [44] was
noted the Konner study while McKeekin et al. [45] colleagues
noted one deep venous thrombosis and one fistula.
In the recurrent setting several trials are being conducted. The OCEANS trial is a randomized study of
carboplatin/gemcitabine and bevacizumab (NCT 00434642)
versus carboplatin/gemcitabine. GOG 213 (Figure 1) is randomized trial in recurrent ovarian cancer patients. Patients
are stratified as to whether or not they are surgical candidates. If the patients are deemed to be surgical candidates
they are randomized to surgery or no surgery followed
by randomization to chemotherapy. If patients are randomized to no surgery they are subsequently randomized
to carboplatin and paclitaxel or carboplatin/paclitaxel and
bevacizumab.
The combination of carboplatin/DOXIL and bevacizumab is also being studied. The later trial may prove
to be intriguing given the recently reported results of the
CALYPSO trial [46]. In the CALYPSO trial the combination of Carboplatin-Doxil demonstrated a superior therapeutic index (benefit/risk ratio) versus current standard,
carboplatin-paclitaxel.
Journal of Oncology
Surgical candidate or not
Surgery
Yes
No
Randomize
Randomize
No surgery
Carboplatin +
paclitaxel
Carboplatin +
paclitaxel +
bevacizumab
Chemo randomization
Figure 1: GOG 213.
3. Small Molecules that Target
the VEGFR Receptor
Small molecule tyrosine kinase inhibitors that target the
vascular endothelial growth factor receptor are currently
being investigated in numerous clinical trials. AZD2171
(Cediranib) is a novel oral tyrosine kinase inhibitor of
VEGFR2, VEGFR1, and c-kit. Matulonis et al. [21] reported
the initial results of this agent in the management of patients
with recurrent ovarian cancer. Five patients had confirmed
partial responses with an overall response rate of 18.5%.
Three patients had stable disease lasting 30, 27+, and 24
weeks. Hirte et al. [22] reported a response rate of 40.5%
in platinum sensitive patients and a response rate of 29%
in platinum resistance disease with AZD 2171 (Cediranib).
Prevalent side effects included fatigue and hypertension.
Currently, ICON-6 is conducting a study of AZD2171
(Cediranib) in platinum sensitive relapsed ovarian cancer in
a three arm randomized placebo-controlled phase III trial in
combination with paclitaxel and carboplatin. (Figure 2).
Pazopanib is tyrosine kinase inhibitor of vascular
endothelial growth factor receptor (VEGFR) −1, −2, and
−3, platelet-derived growth factor receptor (PDGFR) −α
and −β, and c-Kit. Friedlander et al. [24] have reported
activity with pazopanib in women with advanced epithelial
ovarian cancer. Eleven of 36 subjects (31%) experienced a
cancer antigen-125 (CA-125) response to pazopanib. Overall
response rate based on modified Gynecologic Cancer Intergroup (GCIG) criteria (incorporating CA-125, Response
Evaluation Criteria in Solid Tumors (RECIST), and clinical
assessment) was 18% in subjects with measurable disease at
baseline and was 21% in subjects without measurable disease
at baseline. Median PFS was 84 days.
Sunitinib, an inhibitor that targets the VEGFR 1, 2,
3, and platelet–derived growth factor receptors, has also
been studied in the management of patients with recurrent
ovarian cancer. Biagi et al. [25] investigated the role of
sunitinib in the management of patients with recurrent
ovarian cancer. Sunitinib was administered at 50 mg every
Journal of Oncology
3
Table 1: Current trials in ovarian/fallopian/peritoneal cancer.
Author
Burger R et al. [7]
Cannistra et al. [8]
Garcia et al. [9]
Wright et al. [10]
Chura et al. [11]
Nimeiri et al. [12]
Monk et al. [13]
Simpkins et al. [14]
McGonigle et al. [15]
Azad et al. [17]
Micha et al. [16]
Campos/Penson et al. [18, 19]
N
62
44
70
23
15
13
32
25
18
13
20
58
Prior lines
1-2
2-3
1–3
2–15
5–15
1–3
2–10
2–12
0–2
NR
0
0
Platinum S/R∗ /first line
+/+
+/+
+/+
−/+
+/+
+/+
−/+
−/+
−/+
NR
First line
First line
Regimen∗∗
Single
Single
Combo
Combo
Combo
Combo
Single
Combo
Combo
Combo
Combo
Combo
RR:CR + PR
21 %
15.9%
24%
35 %
43 %
15 %
16 %
28 %
22%
46%
80%
75%
TTP/PFS median
PFS 4.7mo
PFS 4.4 mo
TTP 7.2 mo
TTP 5.6 mo
PFS 3.9 mo
PFS 4.1 mo
PFS 5.5 mo
TTP 9.0 mo
PFS 3.8 mo
NR
NR
PFS:11mo
∗ Enrolled
patients: platinum sensitive/resistant/first line.
bevacizumab or combination therapy with cytotoxic or other biological agents.
NR: not reported.
∗∗ Single
Carboplatin/paclitaxel + placebo followed
by placebo for 18 months
Carboplatin/paclitaxel + Cediranib followed
by placebo for 18 months
ICON 6
Randomized
Carboplatin/paclitaxel + Cediranib followed
by Cediranib for 18 months
Figure 2: ICON-6.
day on a 4-week on 2-week off schedule. Noted in this study
was the development of pleural effusions during the 2-week
rest period. Of the seventeen patients that were studied 12%
of patients had a partial response, and 59% of patients had
disease stabilization. Currently the Harvard Cancer Center
Gynecological Group (NCT00768144) is conducting a phase
II trial using sunitinib in refractory ovarian caner patients.
The dose of sunitinib is held constant at 37.5 mg every day.
AMG 706 is an investigational inhibitor of vascular
endothelial growth factor receptors 1, 2, and 3, plateletderived growth factor receptor, and stem-cell factor receptor.
A Phase II Evaluation of AMG706 (NCT00574951) in the
Treatment of Persistent or Recurrent Epithelial Ovarian
Fallopian Tube or Primary Peritoneal Cancer is currently
active.
Matei et al. [26] reported on the activity of sorafenib in
patient with recurrent ovarian cancer. Sorafenib is a tyrosine
kinase inhibitor targeting raf and other receptor kinases
(VEGF-R, PDGF-R, Flt3, c-KIT). Patients received sorafenib
at 400 mg QD. Patients in this study have a 3% partial
response, and 20% of patients had stable disease for > than
6 months. Toxicities included rash, metabolic abnormalities,
gastrointestinal, cardiovascular, and pulmonary toxicity.
4. VEGF Trap (Aflibercept)
VEGF trap (Aflibercept) is fusion protein containing the
VEGF binding regions of both VEGFR-1 and 2 linked
through the Fc region of a human IgG1. Aflibercept binds
VEGF-A and neutralizes all VEGF-A isoforms plus placental
growth factor. This agent is currently being explored in platinum resistant ovarian cancer. Columbo et al. [39] reported
the results of VEGF Trap in patients with symptomatic
malignant ascites. Aflibercept, 4 mg/kg, i.v. was administered
every 2 weeks, in patients with advanced ovarian cancer
and symptomatic ascites requiring frequent paracentesis. Primary endpoint was repeat paracentesis response rate (RPRR)
defined as at least a doubling of time to the first paracentesis
compared to a baseline average. Patients received 1–13 cycles
of aflibercept. The authors reported that the time to the
first paracentesis was 12–205 days. Eight out of ten evaluable
patients achieved a RPRR response as per protocol. Adverse
events included bowel obstruction, nausea, vomiting,
anorexia, edema, and 1 case of bowel perforation. Tew et al.
[40] reported the preliminary results of a randomized phase
II study in patients with recurrent platinum-resistant epithelial ovarian cancer. VEGF Trap was (2 or 4 mg/kg) administered intravenously every 2 weeks in patients with recurrent
4
ovarian cancer was conducted. Five partial responses in a
sample size of 45 patients (11%) were reported.
5. Epidermal Growth Factor Inhibitors
In addition to the VEGF inhibitors, the epidermal growth
factor receptor (EGFR) has emerged as an attractive target
[47–49]. The activation of EGFR signaling pathways is
known to increase proliferation, angiogenesis, and decrease
apoptosis. Several strategies that target the EGFR in gynecologic cancers have included monoclonal antibodies [1–3],
(trastuzumab, pertuzumab, EMD7200) and tyrosine kinases
inhibitors [4–6] (gefitinib, erlotinib, lapatinib and CI-1033).
Bookman and colleagues [1, 50] reported a response rate of
7% in a phase II trial of ovarian cancer patients treated with
trastuzumab. Kaye et al. [51], Amler et al. [52], and Makhija
et al. [53] in independent studies examined pertuzumab, a
humanized recombinant monoclonal antibody that inhibits
the dimerization of HER2 with EGFR, HER 3, and HER4,
in patients with ovarian cancer. As a single agent there
were only modest responses. Gordan et al. [54] recently
published the clinical activity of pertuzumab in advanced
ovarian cancer. There were five partial responses (response
rate 4.3%), eight patients (6.8%) with stable disease lasting
at least 6 months, and 10 patients with CA-125 reduction
of at least 50%. Median progression-free survival (PFS) was
6.6 weeks. Twenty eight percent of the tumor biopsies were
pHER2+ by ELISA. Of note the progression free survival for
pHER2+ patients was 20.9 weeks (n = 8) versus 5.8 weeks for
pHER2−.
Several studies are ongoing. The EORTC have recently
completed a trial investigating erlotinib as maintenance
therapy following first-line chemotherapy in patients with
ovarian cancer (NCT00263822). A phase II open label
trial of erlotinib and bevacizumab is being conducted by
Alberts et al. in patients with advanced ovarian cancer
(NCT00696670).
Unlike other disciplines there is lack of data in the
gynecological literature on who, if any, will benefit from
EGFR inhibitors. Schilder et al. [55] reported that in a sample
size of 55 ovarian cancer patients 3.6% had mutations in
the EGFR tyrosine kinase domain and that the mutation
correlated with a response to gefitinib. Exploratory analyses
in the pertuzumab studies [51–53] suggested that patients
with platinum resistant disease and low levels of HER3
mRNA might benefit from pertuzumab. An additional study
by Tanner et al. [56] demonstrated an influence of HER 3
expression on the survival of patients with ovarian cancer.
Selection of ovarian cancer patients with EGFR amplifications, increased pHER2, and low expression of HER 3
ratios may represent the selected few that may respond to
EGFR inhibitors.
Journal of Oncology
nation of EGFR inhibitors and VEGF inhibitors. Nimeiri
et al. [12] investigated the clinical activity and safety of
bevacizumab and erlotinib patients with recurrent ovarian,
primary peritoneal, and fallopian tube cancer. In this study
patients were heavily pretreated. Two patients had a fatal
bowel perforation.
Currently investigators at the Harvard Cancer Center
are conducting a randomized phase II trial of Bevacizumab or Bevacizumab and Erlotinib as First Line
Consolidation Chemotherapy after Carboplatin, Paclitaxel,
and Bevacizumab (CTA) Induction Therapy for Newly
Diagnosed Advanced Ovarian, Fallopian Tube and Primary
Peritoneal Cancer & Papillary Serous Mullerian Tumors
(NCT00520013) [20].
7. Platelet Derived Growth Factor Inhibitors
Platelet-derived growth factor (PDGF) a prototype for
understanding the function of growth factors and receptor
tyrosine kinases (TK) [58] induces cell growth and survival, transformation, migration, vascular permeability, and
wound healing [59]. PDGF receptor (PDGFR) activation
in cancer occurs as a consequence of gene amplification,
chromosomal rearrangements, or activating mutations [60–
62]. PDGFR activation is critical to tumor initiation in
addition to functioning as a mediator of connective tissue
stroma [63].
PDGFR has been shown in 50–80% of ovarian tumors
[63]. Several agents that target the PDGFR have been studied.
These include imatinib mesylate [63–66], sorafenib, [17, 26],
sunitinib [25], dasatinib [67], 3G3 [68], and CDP 860 [69].
Imatinib mesylate is a selective Abl, c-Kit, and PDGFR
inhibitor. Three phase II clinical trials [64, 70, 71] in patients
with ovarian cancer failed to demonstrate clinical benefit.
The GOG (170 M) is currently studying dasatinib in
a Phase II Evaluation of Dasatinib in the Treatment of
Persistent or Recurrent Epithelial Ovarian, Fallopian Tube,
or Primary Peritoneal Carcinoma
BIBF1120 [72] is a novel agent. It is a triple angiokinase inhibitor that targets the VEGFR, PDGRF, and the
fibroblast growth factor receptor (FGFR). Sustained pathway
inhibition is a distinct feature of this agent. Ledermann et
al. [73] recently conducted a randomized phase II placebocontrolled trial using maintenance therapy to evaluate the
vascular targeting agent BIBF 1120 following treatment of
relapsed ovarian cancer. The 36-week PFS rate for BIBF 1120
was 15.6% and 2.9% for placebo. The authors concluded
that maintenance BIBF 1120 could delay disease progression
in ovarian cancer patients who had previously responded to
chemotherapy.
8. Folate Receptor Inhibitors
6. Combination Therapy with EGFR and
VEGF Inhibitors
EGFR activation has been reported to promote VEGF [57]
secretion. Several clinical studies are exploring the combi-
Folic acid is an essential vitamin and of importance for
one-carbon transfer processes medicated by enzyme systems
involved in DNA synthesis [74]. Increased expression of
α-FR has been described in various tumor tissues, including
ovarian, endometrial, and breast cancer [75]. While the
Journal of Oncology
5
Table 2: PDGF-targeted therapies in ovarian cancer.
clinical trial.gov ID
NCT00913835
NCT00768144
NCT00437372
NCT00792545
NCT00672295
NCT00436215
NCT00526799
NCT00390611
NCT00096200,
NCT00510653
NCT00840450
Therapeutic regimen
Doxil ± IMC 3G3 in platinum refractory or resistant EOC
Sunitinib in refractory/recurrent ovarian, fallopian tube, or peritoneal cancer
Sunitinib and radiation therapy
Dasatinib + bevacizumab in surgically metastatic, or unresectable solid tumors
Dasatinib + paclitaxel + carboplatin in ovarian, fallopian tube, and peritoneal
cancer
Sorafenib + bevacizumab in recurrent/refractory ovarian, fallopian tube, or
peritoneal cancer
Sorafenib + topotecan in platinum resistant EOC
Paclitaxel + carboplatin ± sorafenib for first-line therapy for EOC
Sorafenib + paclitaxel + carboplatin in recurrent platinum-sensitive ovarian,
fallopian tube, or peritoneal cancer
Gleevac study for patients with ovarian cancer
Gleevac and paclitaxel with recurrent mullerian cancers
function of α-FR in cancers is not fully understood,
folates are critical metabolites for nucleotide synthesis and
methylation reactions. Its overexpression might confer a
tumor growth advantage by increasing folate availability to
cancer cells [75]. Over 90% of nonmucinous ovarian cancers
overexpress α-FR [76].
Several strategies have been employed to target the
folate receptor. Some of these include the use of anti-α-FR
antibodies or folic acid conjugates. There has also been recent
research to show that α-FR may have a potential as a target for
immunotherapeutic approaches in ovarian cancer. α-FR is
a tumor-associated antigen that induces detectable immune
responses in 70% of patients with breast and ovarian cancer
[77]. The presence of endogenous immune reactivity raises
the possibility that the immune response could be further
enhanced by vaccines targeting the α-FR. Hernando et al.
[78] presented a case of a women with recurrent epithelial
ovarian cancer treated with a vaccination regimen created
with autologous dendritic cells engineered with mRNAencoded α-FR [78]. An initial contrast-enhanced CT of
the abdomen before vaccination had shown para-aortic
lymph node metastasis at the level of the left renal hilus
and lower abdominal aorta. Follow-up CT 16 months after
last vaccination depicted a more than 50% regression of
lymph node metastasis and a dramatic decrease in CA125
concentrations 4 weeks after the first vaccination [78].
Farletuzumab (MORAb-003) is a monoclonal antibody
to α-FR that activates antibody-dependent cell-mediated
cytotoxicity and complement-mediated toxicity [79]. In a
recent Phase II trial of 54 patients [80] with platinumsensitive relapsed disease patients who received combination
therapy exhibited a prolongation of their remission when
compared to their previous remission. Ongoing clinical
trials looking at Farletuzumab include a Phase III trial
comparing the efficacy and safety of intravenous carboplatin
and taxanes with and without farletuzumab in subjects with
first platinum-sensitive relapse, a Phase II trial examining
intravenous paclitaxel with and without farletuzumab in
patients with first platinum-resistant or refractory relapse.
Study PI
W. McGuire
S. Campos
A . Dicker
E. Kohn
A. Secord
E. Kohn
D. Matei
J. Hainsworth
V. von Gruenigan
D. Gershenson
F. Muggia
EC145 is a drug that is specifically designed to enter
cancer cells via the folate vitamin receptor (FR). Early
clinical evidence in a small number of phase I patients
suggests that EC145 may have antitumor effect in women
with advanced ovarian cancer. Current independent studies
include a study of EC145 in patients with advanced ovarian
and endometrial cancers (NCT00507741) and a study in
patients with platinum resistant ovarian cancer with a
combination of Doxil and EC145 Combination Therapy
(NCT00722592).
9. Poly-ADP-Ribose Polymerase (PARP)
Inhibitors
Between 5 and 10% of all ovarian cancer cases are associated
with inheriting a mutation in the BRCA1 or BRCA2 gene
[81]. The lifetime risk of ovarian cancer for BRCA1 and
BRCA2 mutation carriers is estimated at 40–50% and 10–
20%, respectively. BRCA1 and BRCA2 are essential for the
repair of double strand DNA breaks (DSBs) and maintenance
of genomic stability [82].
Poly (ADP-ribose) polymerase (PARP) is a key nuclear
enzyme involved in the repair of DNA single-strand breaks
(SSBs) using the base excision repair pathway [83]. PARP-1
and PARP-2 are the only members of the PARP family known
to be activated by DNA damage, and PARP-1 has been best
characterized. PARP inhibition results in the accumulation of
DNA SSBs, which may lead to DSBs. Thus, the use of PARP
inhibitors in BRCA mutation carriers uses the concept of
synthetic lethality and hence can be described a therapeutic
exploitation.
In the first human phase I clinical trial using Olaparib (AZD2281, KU-0059436; AstraZeneca) an oral smallmolecule PARP-1 inhibitor, toxicities included nausea, vomiting, anorexia, and fatigue. Efficacy has been reported.
Olaparib has shown antitumor activity in BRCA-associated
ovarian cancer [84, 85]. Fifty patients were treated at various
doses, of which 41 were BRCA1 mutation carriers, eight were
6
Journal of Oncology
Table 3: α-folate receptor inhibitors and ovarian cancer.
Clinical trial.gov ID
NCT00722592
NCT00738699
NCT00849667
Therapeutic regimen
Doxil and EC145in platinum resistant EOC
MORAb-003 in first platinum resistant or refractory relapsed EOC
MORAb-003 in platinum sensitive, first relapse EOC
BRCA2 mutation carriers and one had a compelling family
history for BRCA mutation. Of the 46 patients with evaluable
disease, 41% reached either a complete or partial response.
Eleven percent had meaningful stabilization of disease for
4 months, giving a total clinical benefit rate of 52%. The
median response duration was 30 weeks.
Recently reported were the results of a phase II trial of
the oral PARP inhibitor Olaparib (AZD 2281) in BRCAdeficient advanced ovarian cancer [86]. An international,
phase II study examined two cohorts of patients that received
oral olaparib in 28-day cycles, initially at the MTD, 400 mg
bid (33 pts), and subsequently at 100 mg bid (24 pts). The
confirmed overall response rate was 33% at 400 mg bid dose
and 12.5% at 100 mg bid dose. Clinical benefit rate (ORR
and/or confirmed ≥50% decline in CA125) was 57.6% at
400 mg bid and 16.7% at 100 mg bd. Toxicity was mild.
Olaparib is currently being evaluated in randomized
Phase II trials in platinum-sensitive recurrent ovarian cancer,
and in known BRCA or high grade recurrent ovarian
cancer. It is also being compared with pegylated liposomal
doxorubicin in patients with BRCA mutated ovarian cancer
with a 0-12 month platinum-free interval (NCT00628251).
Other PARP inhibitors are also being evaluated in
BRCA mutation carriers with cancer, including AG0146999
(Pfizer) ABT888 (Abbott), BSI-201 (Bipar), INO-1001
(Inotek/Genentech), and MK4827 (Merck).
The use of PARP inhibitors might be extended to
sporadic ovarian cancers with homologous recombination
defects. These sporadic tumors seem to phenocopy BRCA1/2
deficient tumors even though they do not possess the
germline mutations in either gene. This phenomenon is
called “BRCAness.” This can occur due to loss of heterozygosity, hypermethlyation, and haploinsufficiency (inactivation of one BRCA allele), thereby, genetically silencing the
BRCA gene without an actual germline mutation. A recent
study suggests that over 50% of high-grade serous ovarian
cancer had loss of BRCA function, either by genetic or
epigenetic events [87]. A randomized placebo-controlled
trial of olaparib as a maintenance therapy in patients with
serous (sporadic) ovarian cancer at high risk for recurrence
is now underway.
10. Aurora Kinase Inhibitors
Aurora kinases are protein kinases that are important mitotic
regulators [88, 89]. They are central to many cellular
functions notably mitosis, centromere separation, as well as
mitotic spindle formation. Three aurora kinases (A, B, C)
exist. The activity of aurora kinase is cell cycle dependent and
Study PI
R. Messmann
D. Chakraborty
D. Chakraborty
active during the G2M phase of the cell cycle. Several investigators [88, 90, 91] have described the oncogenic potential
of these proteins. Aurora-A also phosphorylates the tumor
suppressor protein p53, resulting cell cycle progression [92].
Aurora A is overexpressed in 83% of human epithelial
ovarian carcinomas [93]. In addition, amplification of
human chromosome 20q13.2, which contains Aurora-A,
frequently occurs in ovarian cancer [94]. Aurora kinase A has
been significantly associated with tumor grade, FIGO stage,
and survival [93, 95].
Lin et al. [96] studied the role of MK-0457, a small
molecule pan-aurora kinase inhibitor in ovarian cancer cell
models. Two chemosensitive human ovarian cancer cell lines,
HeyA8 and SKOV3ip1, were used to study the effects of
aurora kinase inhibition. Additionally two chemoresistant
cell lines (Hey A8-MDR and A2780-CP-20) were also
studied. Both cell lines showed that aurora kinase inhibition
alone significantly reduced tumor burden. Combination
treatment with docetaxel resulted in significantly improved
reduction in tumor growth beyond that afforded by docetaxel alone (P < or = .03). Scharer et al. [97] also reported
that aurora kinase inhibitors synergize with paclitaxel to
induce apoptosis in ovarian cancer cells.
Manfredl et al. [98] reported the antitumor activity of
MLN8054, an orally active small molecule inhibitor of aurora
kinase. Growth of human tumor xenografts in nude mice was
dramatically inhibited after oral administration of MLN8054
in human tumor xenografts. MLN8054 induced mitotic
accumulation and apoptosis. Given these findings MLN8054
is currently being explored in the management of patients
with platinum-refractory or resistant epithelial, fallopian, or
primary peritoneal carcinoma (NCT00853307).
11. Hedgehog Pathway Inhibitors
Hedgehog signaling plays a role in many processes such as
cell differentiation, growth, and proliferation. This pathway
is active during embryonic development and remains active
in the adult where it is involved in the maintenance of stem
cell populations.
The Hedgehog family [99] has several proteins which
function as signaling molecules. These include Sonic
hedgehog (Shh), Indian hedgehog (Ihh), and Desert
hedgehog (Dhh). There are two receptors that are involved in the Hedgehog pathway. PATCHED1 is a
hedgehog receptor. In the absence of a ligand PATCHED1
inhibits SMOOTHENED, a transmembrane G-coupled
protein. However, when the ligand binds PATCHED1,
SMOOTHENED suppression is relieved resulting in
transcription of the Hedgehog genes. PATCHED1 or
Journal of Oncology
7
Table 4: PARP inhibitors and ovarian cancer.
clinical trial.gov ID
NCT00753545
NCT00679783
NCT00749502
NCT00664781
NCT00647062
Therapeutic regimen
AZD2281 in platinum sensitive EOC
AZD2281 in known BRCA or recurrent EOC
MK4827 in BRCA mutant ovarian cancer
AG014699 in BRCA mutant ovarian cancer
AZD2281 and carboplatin in BRCA mutant EOC
SMOOTHENED receptor mutations or overexpression of
the Hedgehog ligand leads to uncontrolled cell proliferation.
Bhattacharya et al. [99] studied the role of Hedgehog
signaling in ovarian cancer. They utilized a hedgehog pathway blocker and studied the proliferation of ovarian tumors.
They noted that PATCHED1 is downregulated in ovarian
cancer and that this low level expression of the PATCHED1
contributed to the proliferation of ovarian cancer cells. Chen
et al. [100] also examined the expression and the functional
role of the hedgehog signal molecules in ovarian cancer.
They reported that the hedgehog molecules (Shh, Dhh,
Ptch, Smo, and Gli 1 proteins) were increased in malignant
disease. Decreased cell proliferation in ovarian carcinoma
cell lines was observed with Hedgehog pathway inhibitorcyclopamine.
Recently reported was the effect of IPI-926 (Infinity
Pharmaceuticals, Inc., Cambridge, Mass) a novel inhibitor
of the Hedgehog signaling pathway in ovarian cancer grafts.
Data revealed that treatment with cyclopamine, the natural
product of IPI-926 in animals with primary ovarian cancer
grafts, resulted in tumor growth inhibition. This agent is
currently being explored as a Phase I study in patients with
solid tumors (NCT00761696).
Currently recruiting is a study of GDC-0449 (Genentech,
Inc), a Hedgehog pathway inhibitor, as maintenance therapy
in patients with ovarian cancer in a second or third complete
remission. GDC-0449 will be evaluated in approximately 100
patients with ovarian cancer in second or third complete
remission in a randomized, placebo-controlled, doubleblind, multicenter Phase II trial. Patients are randomized
in a 1 : 1 ratio to receive either GDC-0449 or a placebo
comparator and are stratified based on whether their cancer
is in a second or third complete remission. The primary
endpoint of the trial is progression-free survival. Secondary
outcome measures include overall survival, measurement of
Hedgehog ligand expression in archival tissue, and number
and attribution of adverse events.
12. MTOR Inhibitors
Numerous investigators have reported alterations in PTEN
in gynecological malignancies [101]. PTEN is a lipid phosphatase that is associated with cell cycle G1-phase arrest
and apoptosis through the PI3K/AKT/mTOR pathway [102].
The mTOR pathway is a central regulator of cell growth,
proliferation, and apoptosis. The loss of functional PTEN
either through deletion, mutation, or inactivation leads to
the constitutive activation of PI3K effectors in the absence of
exogenous stimuli. Potential therapies targeting the mTOR
Study PI
J. Lederman
K. Gelman
R. Plummer
E. Kohn
pathway include mTOR inhibitors Temsirolimus (CCI-779),
everolimus (RAD001), and deforolimus (AP23573).
In ovarian cancer, AKT activity is frequently elevated
and is closely associated with the upregulation of mTOR
signaling [103]. High levels of AKT activity in vitro result in
hypersensitivity to mTOR inhibitors [103]. An in vivo study
[104] using xenografts of SKOV-3 cells revealed that RAD001
inhibited tumor growth, angiogenesis, and production of
ascites suggesting the potential of mTOR inhibitors in the
treatment of women with ovarian cancer.
GOG trial 170I has recently closed a Phase II Evaluation
of Temsirolimus (CCI-779, mTOR inhibitor) in the Treatment of Persistent or Recurrent Epithelial Ovarian, Fallopian
Tube or Primary Peritoneal Carcinoma. Currently recruiting
studies include (NCT00926107), a study of the mTOR
inhibitor Temsirolimus (CCI-779) to treat ovarian cancer
with Ca125 relapse only, a Phase I study of DOXIL and Temsirolimus in Resistant Solid Malignancies NCT00703170, and
a Phase I study of Docetaxel and Temsirolimus in resistant
solid malignancies (NCT00703625).
13. Conclusion
Multiple attractive targets for the design of targeted therapeutics in ovarian cancer are currently under investigation. Recent studies employing monoclonal antibodies
have revealed improvements in time to progression. Studies
with tyrosine kinases inhibitors remain in their infancy of
development but have provided the basis for continued
research.
Despite these advances there are multiple goals for
the future. These include a better understanding of the
redundant pathways that exist in cell signaling, creative
targeting of horizontal and vertical signaling pathways,
identification of other predictive markers to better identify
a targeted subpopulation of patients that will respond, and
an underlying of the mechanisms of resistance. Achieving
these goals will be of paramount importance in the study of
targeted therapy in ovarian cancer.
References
[1] M. A. Bookman, K. M. Darcy, D. Clarke-Pearson, R. A.
Boothby, and I. R. Horowitz, “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,” Journal of Clinical
Oncology, vol. 21, no. 2, pp. 283–290, 2003.
8
[2] M. S Gordon, D. Matei, C. Aghajanian, et al., “Clinical activity of pertuzumab (rhuMab 2C4) in advanced, refractory
or recurrent ovarian cancer (OC), and the role of HER2
activation status,” Journal of Clinical Oncology, vol. 23, no.
16S, 2005, abstract 5051.
[3] M. V. Seiden, H. A. Burris, U. Matulonis, et al., “A phase II
trial of EMD72000 (matuzumab), a humanized anti-EGFR
monoclonal antibody, in patients with platinum-resistant
ovarian and primary peritoneal malignancies,” Gynecologic
Oncology, vol. 104, no. 3, pp. 727–731, 2007.
[4] E. M. Posadas, M. S. Liel, V. Kwitkowski, et al., “A phase
II and pharmacodynamic study of gefitinib in patients with
refractory or recurrent epithelial ovarian cancer,” Cancer, vol.
109, no. 7, pp. 1323–1330, 2007.
[5] A. N. Gordon, N. Finkler, R. P. Edwards, et al., “Efficacy
and safety of erlotinib HCl, an epidermal growth factor
receptor (HER1/EGFR) tyrosine kinase inhibitor, in patients
with advanced ovarian carcinoma: results from a phase
II multicenter study,” International Journal of Gynecological
Cancer, vol. 15, no. 5, pp. 785–792, 2005.
[6] S. Campos, O. Hamid, M. V. Seiden, et al., “Multicenter, randomized phase II trial of oral CI-1033 for previously treated
advanced ovarian cancer,” Journal of Clinical Oncology, vol.
23, no. 24, pp. 5597–5604, 2005.
[7] R. A. Burger, M. W. Sill, B. J. Monk, B. E. Greer, and J.
I. Sorosky, “Phase II trial of bevacizumab in persistent or
recurrent epithelial ovarian cancer or primary peritoneal
cancer: a Gynecologic Oncology Group study,” Journal of
Clinical Oncology, vol. 25, no. 33, pp. 5165–5171, 2007.
[8] S. A. Cannistra, U. A. Matulonis, R. T. Penson, et al.,
“Phase II study of bevacizumab in patients with platinumresistant ovarian cancer or peritoneal serous cancer,” Journal
of Clinical Oncology, vol. 25, no. 33, pp. 5180–5186, 2007.
[9] A. A. Garcia, H. Hirte, G. Fleming, et al., “Phase II
clinical trial of bevacizumab and low-dose metronomic oral
cyclophosphamide in recurrent ovarian cancer: a trial of the
California, Chicago, and Princess Margaret Hospital Phase II
Consortia,” Journal of Clinical Oncology, vol. 26, no. 1, pp.
76–82, 2008.
[10] J. D. Wright, A. Hagemann, J. S. Rader, et al., “Bevacizumab
combination therapy in recurrent, platinum-refractory,
epithelial ovarian carcinoma,” Cancer, vol. 107, no. 1, pp. 83–
89, 2006.
[11] J. C. Chura, K. Van Iseghem, L. S. Downs Jr., L. F. Carson,
and P. L. Judson, “Bevacizumab plus cyclophosphamide in
heavily pretreated patients with recurrent ovarian cancer,”
Gynecologic Oncology, vol. 107, no. 2, pp. 326–330, 2007.
[12] H. S. Nimeiri, A. M. Oza, R. J. Morgan, et al., “Efficacy
and safety of bevacizumab plus erlotinib for patients with
recurrent ovarian, primary peritoneal, and fallopian tube
cancer: a trial of the Chicago, PMH, and California Phase II
Consortia,” Gynecologic Oncology, vol. 110, no. 1, pp. 49–55,
2008.
[13] B. J. Monk, E. Han, C. A. Josephs-Cowan, G. Pugmire, and
R. A. Burger, “Salvage bevacizumab (rhuMAB VEGF)-based
therapy after multiple prior cytotoxic regimens in advanced
refractory epithelial ovarian cancer,” Gynecologic Oncology,
vol. 102, no. 2, pp. 140–144, 2006.
[14] F. Simpkins, J. L. Belinson, P. G. Rose, et al., “Avoiding
bevacizumab related gastrointestinal toxicity for recurrent
ovarian cancer by careful patient screening,” Gynecologic
Oncology, vol. 107, no. 1, pp. 118–123, 2007.
[15] K. F. McGonigle, H. G. Muntz, J. L. Vuky, et al., “A phase
II prospective study of weekly topotecan and bevacizumab
Journal of Oncology
[16]
[17]
[18]
[19]
[20]
[21]
[22]
[23]
[24]
[25]
[26]
[27]
[28]
[29]
in platinum refractory ovarian cancer or peritoneal cancer,”
Journal of Clinical Oncology, vol. 26, 2008, abstract 5551.
J. P. Micha, B. H. Goldstein, M. A. Rettenmaier, et al., “A
phase II study of outpatient first-line paclitaxel, carboplatin,
and bevacizumab for advanced-stage epithelial ovarian,
peritoneal, and fallopian tube cancer,” International Journal
of Gynecological Cancer, vol. 17, no. 4, pp. 771–776, 2007.
N. S. Azad, E. M. Posadas, V. E. Kwitkowski, et al., “Combination targeted therapy with sorafenib and bevacizumab
results in enhanced toxicity and antitumor activity,” Journal
of Clinical Oncology, vol. 26, no. 22, pp. 3709–3714, 2008.
R. T. Penson, S. Cannistra, M. V. Seiden, et al., “Phase II
study of carboplatin, paclitaxel and bevacizumab as first line
therapy and consolidation for advanced mullerian tumors,”
Journal of Clinical Oncology, vol. 24, no. 18S, 2006, abstract
5020.
S. C. Campos, D. S. Dizon, S. A. Cannistra, et al., “Safety
of maintenance bevacizumab after first-line chemotherapy
for advanced ovarian mullerian cancers,” Journal of Clinical
Oncology, vol. 25, no. 18S, 2007, abstract 5517.
S. M. Campos, R. T. Penson, U. Matulonis, et al., “STAC: a
randomized phase II trial of avastin or avastin + erlotinib as
first line consolidation chemotherapy after standard therapy,”
British Gynecological Society Meeting, 2008, abstract 93.
U. A. Matulonis, S. Berlin, P. Ivy, et al., “Cediranib, an
oral inhibitor of vascular endothelial growth factor receptor
kinases, is an active drug in recurrent epithelial ovarian,
fallopian tube and peritoneal cancer,” Journal of Clinical
Oncology, vol. 27, no. 33, pp. 5601–5606, 2009.
H. W. Hirte, L. Vidal, G. F. Fleming, et al., “A phase II study
of cediranib (AZD2171) in recurrent or persistent ovarian,
peritoneal or fallopian tube cancer. Final results of a PMH,
Chicago and California consortia trial,” Journal of Clinical
Oncology, vol. 26, no. 298S, 2008, abstract 5521.
R. Buckstein, R. M. Meyer, L. Seymour, et al., “Phase II testing
of sunitinib: the National Cancer Institute of Canada Clinical
Trials Group IND Program Trials IND. 182–185,” Current
Oncology, vol. 14, no. 4, pp. 154–161, 2007.
M. Friedlander, K. C. Hancock, B. Benigno, et al., “Pazopanib
(GW 786034) is active in women with advanced epithelial
ovarian, fallopian tube, and peritoneal cancers: results of a
phase II study,” Journal of Clinical Oncology, vol. 25, no. 18S,
2007, abstract 5561.
J. J. Biagi, A. M. Oza, R. Grimshaw, et al., “A phase II
study of sunitinib (SU11248) in patients with recurrent
epithelial ovarian cancer, fallopian tube or primary peritoneal carcinoma-NCIC CTG IND 185,” Journal of Clinical
Oncology, vol. 26, 2008, abstract 5522.
D. Matei, M. W. Still, K. DeGeest, et al., “Phase II study of
sorafenib in persistent or recurrent epithelial ovarian cancer(
EOC) or primary peritoneal cancer ( PPC): a Gynecology
Oncology Group (GOG) study,” Journal of Clinical Oncology,
vol. 26, 2008, abstract 5537.
E. J. Battegay, “Angiogenesis: mechanistic insights, neovascular diseases, and therapeutic prospects,” Journal of Molecular
Medicine, vol. 73, no. 7, pp. 333–346, 1995.
J. Folkman, “Angiogenesis in cancer, vascular, rheumatoid
and other disease,” Nature Medicine, vol. 1, no. 1, pp. 27–31,
1995.
H. C. Hollingsworth, E. C. Kohn, S. M. Steinberg, M. L.
Rothenberg, and M. J. Merino, “Tumor angiogenesis in
advanced stage ovarian carcinoma,” The American Journal of
Pathology, vol. 147, no. 1, pp. 33–41, 1995.
Journal of Oncology
[30] H. F. Dvorak, “Vascular permeability factor/vascular
endothelial growth factor: a critical cytokine in tumor
angiogenesis and a potential target for diagnosis and
therapy,” Journal of Clinical Oncology, vol. 20, no. 21, pp.
4368–4380, 2002.
[31] J. Folkman, “Tumor angiogenesis: therapeutic implications,”
The New England Journal of Medicine, vol. 285, no. 21, pp.
1182–1186, 1971.
[32] N. Ferrara, “Role of vascular epidermal growth factor in
regulation of angiogenesis,” Kidney International, vol. 56, no.
3, pp. 794–814, 1995.
[33] A. A. Alvarez, H. R. Krigman, R. S. Whitaker, R. K.
Dodge, and G. C. Rodriguez, “The prognostic significance
of angiogenesis in epithelial ovarian carcinoma,” Clinical
Cancer Research, vol. 5, no. 3, pp. 587–591, 1999.
[34] G. Gasparini, E. Bonoldi, G. Viale, et al., “Prognostic
and predictive value of tumour angiogenesis in ovarian
carcinomas,” International Journal of Cancer, vol. 69, no. 3,
pp. 205–211, 1996.
[35] G. H. Shen, M. Ghazizadeh, O. Kawanami, et al., “Prognostic
significance of vascular endothelial growth factor expression
in human ovarian carcinoma,” British Journal of Cancer, vol.
83, no. 2, pp. 196–203, 2000.
[36] M. Sönmezer, M. Güngör, A. Ensari, and F. Ortaç, “Prognostic significance of tumor angiogenesis in epithelial ovarian
cancer: in association with transforming growth factor β and
vascular endothelial growth factor,” International Journal of
Gynecological Cancer, vol. 14, no. 1, pp. 82–88, 2004.
[37] P. J. Paley, K. A. Staskus, K. Gebhard, et al., “Vascular
endothelial growth factor expression in early stage ovarian
carcinoma,” Cancer, vol. 80, no. 1, pp. 98–106, 1997.
[38] E. M. Hartenbach, T. A. Olson, J. J. Goswitz, et al., “Vascular
endothelial growth factor (VEGF) expression and survival
in human epithelial ovarian carcinomas,” Cancer Letters, vol.
121, no. 2, pp. 169–175, 1997.
[39] N. Colombo, G. Mangili, S. Mammoliti, et al., “Aflibercept
(VEGF Trap) for advanced epithelial ovarian cancer (EOC)
patients (pts) with symptomatic malignant ascites: preliminary results of a pilot study,” Journal of Clinical Oncology, vol.
26, 2008, abstract 14598.
[40] W. Tew, N. Colombo, I. Ray-Caquard, et al., “VEGF-Trap for
patients with recurrent platinum resistant epithelial ovarian
cancer,” Journal of Clinical Oncology, vol. 25, no. 18S, 2008,
abstract 5508.
[41] D. S. Alberts, P. Y. Liu, E. V. Hannigan, et al., “Intraperitoneal cisplatin plus intravenous cyclophosphamide versus
intravenous cisplatin plus intravenous cyclophosphamide
for stage III ovarian cancer,” The New England Journal of
Medicine, vol. 335, no. 26, pp. 1950–1955, 1996.
[42] M. Markman, B. N. Bundy, D. S. Alberts, 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,” Journal of Clinical
Oncology, vol. 19, no. 4, pp. 1001–1007, 2001.
[43] D. K. Armstrong, B. Bundy, L. Wenzel, et al., “Intraperitoneal
cisplatin and paclitaxel in ovarian cancer,” The New England
Journal of Medicine, vol. 354, no. 1, pp. 34–43, 2006.
[44] J. A. Konner, D. Grabon, S. Pezzulli, et al., “A phase II study
of intravenous (IV) and intraperitoneal (IP) paclitaxel, IP
cisplatin, and IV bevacizumab as first-line chemotherapy
9
[45]
[46]
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
for optimal stage II or III ovarian, primary peritoneal, and
fallopian tube cancer,” Journal of Clinical Oncology, vol. 27,
no. 15S, 2009, abstract 5539.
D. D. McMeekin, G. Lanneau, J. Curiel, et al., “Phase
II study of intravenous (IV) bevacizumab and paclitaxel,
and intraperitoneal (IP) cisplatin, followed by bevacizumab
consolidation for advanced ovarian (O) or peritoneal (P)
cancers,” Journal of Clinical Oncology, vol. 27, no. 15S, 2009,
abstract 5540.
E. Pujade-Lauraine, S. Mahner, J. Kaern, et al., “A randomized, phase III study of carboplatin and pegylated liposomal
doxorubicin versus carboplatin and paclitaxel in relapsed
platinum-sensitive ovarian cancer (OC): CALYPSO study
of the Gynecologic Cancer Intergroup (GCIG),” Journal of
Clinical Oncology, vol. 27, no. 18S, 2009, abstract LBA5509.
J. Mendelsohn and J. Baselga, “The EGF receptor family as
targets for cancer therapy,” Oncogene, vol. 19, no. 56, pp.
6550–6565, 2000.
B. F. El-Rayes and P. M. LoRusso, “Targeting the epidermal
growth factor receptor,” British Journal of Cancer, vol. 91, no.
3, pp. 418–424, 2004.
R. Nahta and F. J. Esteva, “HER-2-targeted therapy: lessons
learned and future directions,” Clinical Cancer Research, vol.
9, no. 14, pp. 5078–5084, 2003.
M. A. Bookman, “Biological therapy of ovarian cancer:
current directions,” Seminars in Oncology, vol. 25, no. 3, pp.
381–396, 1998.
S. B. Kaye, C. J. Poole, M. Bidzinksi, et al., “A randomized
phase II study evaluating the combination of carboplatinbased chemotherapy with pertuzumab versus carboplatin
based chemotherapy in patients with relapsed platinum
sensitive ovarian cancer,” Journal of Clinical Oncology, vol. 26,
2008, abstract 5520.
L. Amler, S. Makhija, and T. Januario, “Her 2 pathway gene
expression analysis in a phase II study of pertuzumab+
gemcitabine vs. gemcitabine + placebo in patients with
platinum resistant epithelial ovarian cancer,” Journal of
Clinical Oncology, vol. 26, 2008, abstract 5562.
S. Makhija, D. Glenn, F. Ueland, et al., “Results from a
phase II randomized, placebo-controlled, double-blind trial
suggest improved PFS with the addition of pertuzumab
to gemcitabine in patients with platinum-resistant ovarian,
fallopian tube, or primary peritoneal cancer,” Journal of
Clinical Oncology, vol. 25, no. 18S, 2007, abstract 5507.
M. S. Gordon, D. Matei, C. Aghajanian, et al., “Clinical
activity of pertuzumab (rhuMAb2C4), a HER dimerization
inhibitor, in advanced ovarian cancer: potential predicative
relationship between tumor HER 2 activation status,” Journal
of Clinical Oncology, vol. 24, no. 26, pp. 4324–4332, 2006.
R. J. Schilder, M. V. Sill, W. Chen, et al., “Phase II study
of Getinib in patients with relapsed or persistent ovarian or
primary peritoneal carcinoma and evaluation of epidermal
growth factor receptor mutations and immunohistochemical
expression: a Gynecological Oncology Group Study,” Clinical
Cancer Research, vol. 11, no. 15, pp. 5539–5548, 2005.
B. Tanner, D. Hasenclever, K. Stern, et al., “ErbB-3 predicts
survival in ovarian cancer,” Journal of Clinical Oncology, vol.
24, no. 26, pp. 4317–4323, 2006.
R. S. Herbst, D. H. Johnson, E. Mininberg, et al., “Phase I/II
trial evaluating the anti-vascular endothelial growth factor
monoclonal antibody bevacizumab in combination with
the HER-1/epidermal growth factor receptor tyrosine kinase
inhibitor erlotinib for patients with recurrent non-small-cell
10
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
[68]
[69]
[70]
[71]
Journal of Oncology
lung cancer,” Journal of Clinical Oncology, vol. 23, no. 11, pp.
2544–2555, 2005.
J. Andrae, R. Gallini, and C. Betsholtz, “Role of plateletderived growth factors in physiology and medicine,” Genes
& Development, vol. 22, no. 10, pp. 1276–1312, 2008.
J. Schmitt and D. Matei, “Platelet-derived growth factor
pathway inhibitors in ovarian cancer,” Clinical Ovarian
Cancer, vol. 1, no. 2, pp. 120–126, 2008.
X. Mao and R. A. Hamoudi, “Molecular and cytogenetic
analysis of glioblastoma multiforme,” Cancer Genetics and
Cytogenetics, vol. 122, no. 2, pp. 87–92, 2000.
M. Carroll, M. H. Tomasson, G. F. Barker, T. R. Golub,
and D. G. Gilliland, “The TEL/platelet-derived growth factor
β receptor (PDGFβR) fusion in chronic myelomonocytic
leukemia is a transforming protein that self-associates and
activates PDGFβR kinase-dependent signaling pathways,”
Proceedings of the National Academy of Sciences of the United
States of America, vol. 93, no. 25, pp. 14845–14850, 1996.
M. C. Heinrich, C. L. Corless, A. Duensing, et al., “PDGFRA
activating mutations in gastrointestinal stromal tumors,”
Science, vol. 299, no. 5607, pp. 708–710, 2003.
S. M. Apte, C. D. Bucana, J. J. Killion, D. M. Gershenson,
and I. J. Fidler, “Expression of platelet-derived growth factor
and activated receptor in clinical specimens of epithelial
ovarian cancer and ovarian carcinoma cell lines,” Gynecologic
Oncology, vol. 93, no. 1, pp. 78–86, 2004.
R. L. Coleman, R. R. Broaddus, D. C. Bodurka, et al.,
“Phase II trial of imatinib mesylate in patients with recurrent platinum- and taxane-resistant epithelial ovarian and
primary peritoneal cancers,” Gynecologic Oncology, vol. 101,
no. 1, pp. 126–131, 2006.
E. M. Posadas, V. Kwitkowski, H. L. Kotz, et al., “A prospective
analysis of imatinib-induced c-KIT modulation in ovarian
cancer: a phase II clinical study with proteomic profiling,”
Cancer, vol. 110, no. 2, pp. 309–317, 2007.
D. Matei, D. D. Chang, and M.-H. Jeng, “Imatinib mesylate
(Gleevac) inhibits ovarian cancer cell growth through a
mechanisms dependent on platelet derived growth factor α
and Akt inactivation,” Clinical Cancer Research, vol. 10, no. 2,
pp. 681–690, 2004.
E. Buchdunger, J. Zimmermann, H. Mett, et al., “Selective inhibition of the platelet-derived growth factor signal
transduction pathway by a protein-tyrosine kinase inhibitor
of the 2-phenylamniopyridmidine class,” Proceedings of the
National Academy of Sciences of the United States of America,
vol. 92, no. 7, pp. 2558–2562, 1995.
H. Youssoufian, R. J. Amato, C. J. Sweeney, et al., “Phase I
study of IMC-3G3, and IgG1 monoclonal antibody targeting
PDGFRα in patients with advanced solid malignancies,”
Journal of Clinical Oncology, vol. 26, no. 636S, 2008.
G. C. Jayson, G. J. M. Parker, S. Mullamitha, et al., “Blockade
of platelet-derived growth factor receptor-beta by CDP860,
a humanized, PEGylated di-Fab’, leads to fluid accumulation
and is associated with increased tumor vascularized volume,”
Journal of Clinical Oncology, vol. 23, no. 5, pp. 973–981, 2005.
R. J. Schilder, M. W. Sill, R. B. Lee, et al., “Phase II evaluation
of imatinib mesylate in the treatment of recurrent or persistent epithelial ovarian or primary peritoneal carcinoma:
a Gynecologic Oncology Group study,” Journal of Clinical
Oncology, vol. 26, no. 20, pp. 3418–3425, 2008.
D. S. Alberts, P. Y. Liu, S. P. Wilczynski, et al., “Phase II trial of
imatinib mesylate in recurrent, biomarker positive, ovarian
cancer,” International Journal of Gynecological Cancer, vol. 17,
no. 4, pp. 784–788, 2007.
[72] F. Hilberg, G. J. Roth, M. Krssak, et al., “BIBF 1120: triple
angiokinase inhibitor with sustained receptor blockade and
good antitumor efficacy,” Cancer Research, vol. 68, no. 12, pp.
4774–4782, 2008.
[73] J. A. Ledermann, G. J. Rustin, A. Hackshaw, et al., “A
randomized phase II placebo-controlled trial using maintenance therapy to evaluate the vascular targeting agent BIBF
1120 following treatment of relapsed ovarian cancer (OC),”
Journal of Clinical Oncology, vol. 27, no. 15S, 2009, abstract
5501.
[74] S. Markert, S. Lassmann, B. Gabriel, et al., “Alpha-folate
receptor expression in epithelial ovarian carcinoma and nonneoplastic ovarian tissue,” Anticancer Research, vol. 28, no. 6,
pp. 3567–3572, 2008.
[75] H. Elnakat and M. Ratnam, “Role of folate receptor genes in
reproduction and related cancers,” Frontiers in Bioscience, vol.
11, pp. 506–519, 2006.
[76] K. R. Kalli, A. L. Oberg, G. L. Keeney, et al., “Folate
receptor alpha as a tumor target in epithelial ovarian cancer,”
Gynecologic Oncology, vol. 108, pp. 619–626, 2008.
[77] K. L. Knutson, C. J. Krco, C. L. Erskine, et al., “T-cell
immunity to the folate receptor alpha is prevalent in women
with breast or ovarian cancer,” Journal of Clinical Oncology,
vol. 24, no. 26, pp. 4254–4261, 2006.
[78] J. J. Hernando, T.-W. Park, H.-P. Fischer, et al., “Vaccination
with dendritic cells transfected with mRNA-encoded folatereceptor-α for relapsed metastatic ovarian cancer,” The Lancet
Oncology, vol. 8, no. 5, pp. 451–454, 2007.
[79] W. Ebel, E. L. Routhier, B. Foley, et al., “Preclinical evaluation
of MORAb-003, a humanized monoclonal antibody antagonizing folate receptor-alpha,” Cancer Immunity, vol. 7, pp.
6–13, 2007.
[80] D. K. Armstrong, A. Bicher, R. L. Coleman, et al.,
“Exploratory phase II efficacy study of MORAb-003, a monoclonal antibody against folate receptor alpha, in platinumsensitive ovarian cancer in first relapse,” Journal of Clinical
Oncology, vol. 26, 2008, abstract 5500.
[81] D. Ford, D. F. Easton, D. T. Bishop, S. A. Narod, and D. E.
Goldgar, “Risks of cancer in BRCA1-mutation carriers,” The
Lancet, vol. 343, no. 8899, pp. 692–695, 1994.
[82] C. J. Lord and A. Ashworth, “Targeted therapy for cancer
using PARP inhibitors,” Current Opinion in Pharmacology,
vol. 8, no. 4, pp. 363–369, 2008.
[83] A. N. J. Tutt, C. J. Lord, N. McCabe, et al., “Exploiting the
DNA repair defect in BRCA mutant cells in the design of
new therapeutic strategies for cancer,” Cold Spring Harbor
Symposia on Quantitative Biology, vol. 70, pp. 139–148, 2005.
[84] P. C. Fong, D. S. Boss, C. P. Carden, et al., “AZD2281 [KU0059436], a PARP [poly ADP-ribose polymerase] inhibitor
with single agent anticancer activity in patients with BRCA
deficient ovarian cancer: results from a phase I study,” Journal
of Clinical Oncology, vol. 26, 2008, abstract 5510.
[85] C. Carden, et al., “Phase I study of AZD2281 [KU-0059436],
a PARP [poly ADP-ribose polymerase] inhibitor: results in
patients with BRCA deficient ovarian cancer,” in Proceedings
of the National Cancer Research Institute Cancer Conference,
vol. 320, 2008, abstract BOA22.
[86] M. W. Audeh, R. T. Penson, M. Friedlander, et al., “Phase
II trial of the oral PARP inhibitor olaparib (AZD2281) in
BRCA-deficient advanced ovarian cancer,” Journal of Clinical
Oncology, vol. 27, no. 15S, 2009, abstract 5500.
[87] J. Z. Press, A. De Luca, N. Boyd, et al., “Ovarian carcinomas
with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities,” BMC Cancer, vol. 8, article 17, 2008.
Journal of Oncology
[88] E. A. Harrington, D. Bebbington, J. Moore, et al., “VX-680,
a potent and selective small-molecule inhibitor of the Aurora
kinases, suppresses tumor growth in vivo,” Nature Medicine,
vol. 10, no. 3, pp. 262–267, 2004.
[89] N. Keen and S. Taylor, “Aurora-kinase inhibitors as anticancer agents,” Nature Reviews Cancer, vol. 4, no. 12, pp. 927–
936, 2004.
[90] N. Matthews, C. Visintin, B. Hartzoulakis, A. Jarvis, and D.
L. Selwood, “Aurora A and B kinases as targets for cancer:
will they be selective for tumors?” Expert Review of Anticancer
Therapy, vol. 6, no. 1, pp. 109–120, 2006.
[91] H. Zhou, J. Kuang, L. Zhong, et al., “Tumour amplified
kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation,” Nature Genetics, vol. 20, no. 2,
pp. 189–193, 1998.
[92] H. Katayama, K. Sasai, H. Kawai, et al., “Phosphorylation by
aurora kinase A induces Mdm2-mediated destabilization and
inhibition of p53,” Nature Genetics, vol. 36, no. 1, pp. 55–62,
2004.
[93] C. N. Landen Jr., Y. G. Lin, A. Immaneni, et al., “Overexpression of the centrosomal protein Aurora-A kinase is associated
with poor prognosis in epithelial ovarian cancer patients,”
Clinical Cancer Research, vol. 13, no. 14, pp. 4098–4104, 2007.
[94] M. M. Tanner, S. Grenman, A. Koul, et al., “Frequent
amplification of chromosomal region 20q12-q13 in ovarian
cancer,” Clinical Cancer Research, vol. 6, no. 5, pp. 1833–1839,
2000.
[95] A. A. Kulkarni, M. Loddo, E. Leo, et al., “DNA replication
licensing factors and aurora kinases are linked to aneuploidy
and clinical outcome in epithelial ovarian carcinoma,” Clinical Cancer Research, vol. 13, no. 20, pp. 6153–6161, 2007.
[96] Y. G. Lin, A. Immaneni, W. M. Merritt, et al., “Targeting Aurora kinase with MK-0457 inhibits ovarian cancer
growth,” Clinical Cancer Research, vol. 14, no. 17, pp. 5437–
5446, 2008.
[97] C. D. Scharer, N. Laycock, A. O. Osunkoya, et al., “Aurora
kinase inhibitors synergize with paclitaxel to induce apoptosis in ovarian cancer cells,” Journal of Translational Medicine,
vol. 6, article 79, 2008.
[98] M. G. Manfredi, J. A. Ecsedy, K. A. Meetze, et al., “Antitumor activity of MLN8054, an orally active small-molecule
inhibitor of Aurora a kinase,” Proceedings of the National
Academy of Sciences of the United States of America, vol. 104,
no. 10, pp. 4106–4111, 2007.
[99] R. Bhattacharya, J. Kwon, B. Ali, et al., “Role of hedgehog
signaling in ovarian cancer,” Clinical Cancer Research, vol. 14,
no. 23, pp. 7659–7666, 2008.
[100] X. Chen, A. Horiuchi, N. Kikuchi, et al., “Hedgehog signal
pathway is activated in ovarian carcinomas, correlating with
cell proliferation: it’s inhibition leads to growth suppression
and apoptosis,” Cancer Science, vol. 98, no. 1, pp. 68–76, 2007.
[101] B. H. Jiang and L. Z. Liu, “PI3K/PTEN signaling in tumorgenesis and angiogenesis,” Biochimica et Biophysica Acta, vol.
1784, no. 1, pp. 150–158, 2008.
[102] I. Sansal and W. R. Sellers, “The biology and clinical relevance
of the PTEN tumor suppressor pathway,” Journal of Clinical
Oncology, vol. 22, no. 14, pp. 2954–2963, 2004.
[103] J .F. Gera, I. K. Mellinghoff, Y. Shi, et al., “AKT activity
determines sensitivity to mammalian target of rapamycin
(mTOR) inhibitors by regulating cyclin D and c-myc expression,” The Journal of Biological Chemistry, vol. 279, no. 4, pp.
2737–2746, 2004.
[104] S. Mabuchi, D. A. Altomare, M. Cheung, et al., “RAD001
inhibits human ovarian cancer cell proliferation, enhances
11
cisplatin-induced apoptosis, and prolongs survival in an
ovarian cancer model,” Clinical Cancer Research, vol. 13, no.
14, pp. 4261–4270, 2007.
MEDIATORS
of
INFLAMMATION
The Scientiic
World Journal
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Gastroenterology
Research and Practice
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Journal of
Diabetes Research
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
International Journal of
Journal of
Endocrinology
Immunology Research
Hindawi Publishing Corporation
http://www.hindawi.com
Disease Markers
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Volume 2014
Submit your manuscripts at
http://www.hindawi.com
BioMed
Research International
PPAR Research
Hindawi Publishing Corporation
http://www.hindawi.com
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Volume 2014
Journal of
Obesity
Journal of
Ophthalmology
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Evidence-Based
Complementary and
Alternative Medicine
Stem Cells
International
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Journal of
Oncology
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Parkinson’s
Disease
Computational and
Mathematical Methods
in Medicine
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
AIDS
Behavioural
Neurology
Hindawi Publishing Corporation
http://www.hindawi.com
Research and Treatment
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014
Oxidative Medicine and
Cellular Longevity
Hindawi Publishing Corporation
http://www.hindawi.com
Volume 2014