ArticlePDF Available

Current status and implications of microRNAs in ovarian cancer diagnosis and therapy

Authors:

Abstract and Figures

Ovarian cancer is the fifth most common cancer among women and causes more deaths than any other type of female reproductive cancer. Currently, treatment of ovarian cancer is based on the combination of surgery and chemotherapy. While recurrent ovarian cancer responds to additional chemotherapy treatments, the progression-free interval becomes shorter after each cycle, as chemo-resistance increases until the disease becomes incurable. There is, therefore, a strong need for prognostic and predictive markers to help optimize and personalize treatment in order to improve the outcome of ovarian cancer. An increasing number of studies indicate an essential role for microRNAs in ovarian cancer progression and chemo-resistance. MicroRNAs (miRNAs) are small endogenous non-coding RNAs (~22bp) which are frequently dysregulated in cancer. Typically, miRNAs are involved in crucial biological processes, including development, differentiation, apoptosis and proliferation. Two families of miRNAs, miR-200 and let-7, are frequently dysregulated in ovarian cancer and have been associated with poor prognosis. Both have been implicated in the regulation of epithelial-to-mesenchymal transition, a cellular transition associated with tumor aggressiveness, tumor invasion and chemo-resistance. Moreover, miRNAs also have possible implications for improving cancer diagnosis; for example miR-200 family, let-7 family, miR-21 and miR-214 may be useful in diagnostic tests to help detect ovarian cancer at an early stage. Additionally, the use of multiple target O-modified antagomirs (MTG-AMO) to inhibit oncogenic miRNAs and miRNA replacement therapy for tumor suppressor miRNAs are essential tools for miRNA based cancer therapeutics. In this review we describe the current status of the role miRNAs play in ovarian cancer and focus on the possibilities of microRNA-based therapies and the use of microRNAs as diagnostic tools.
Content may be subject to copyright.
R E V I E W Open Access
Current status and implications of microRNAs in
ovarian cancer diagnosis and therapy
Mohd Saif Zaman
1
, Diane M Maher
1
, Sheema Khan
1
, Meena Jaggi
1,2,3
and Subhash C Chauhan
1,2,3*
Abstract
Ovarian cancer is the fifth most common cancer among women and causes more deaths than any other type of
female reproductive cancer. Currently, treatment of ovarian cancer is based on the combination of surgery and
chemotherapy. While recurrent ovarian cancer responds to additional chemotherapy treatments, the
progression-free interval becomes shorter after each cycle, as chemo-resistance increases until the disease becomes
incurable. There is, therefore, a strong need for prognostic and predictive markers to help optimize and personalize
treatment in order to improve the outcome of ovarian cancer. An increasing number of studies indicate an
essential role for microRNAs in ovarian cancer progression and chemo-resistance. MicroRNAs (miRNAs) are small
endogenous non-coding RNAs (~22bp) which are frequently dysregulated in cancer. Typically, miRNAs are involved
in crucial biological processes, including development, differentiation, apoptosis and proliferation. Two families of
miRNAs, miR-200 and let-7, are frequently dysregulated in ovarian cancer and have been associated with poor
prognosis. Both have been implicated in the regulation of epithelial-to-mesenchymal transition, a cellular transition
associated with tumor aggressiveness, tumor invasion and chemo-resistance. Moreover, miRNAs also have possible
implications for improving cancer diagnosis; for example miR-200 family, let-7 family, miR-21 and miR-214 may be
useful in diagnostic tests to help detect ovarian cancer at an early stage. Additionally, the use of multiple target
O-modified antagomirs (MTG-AMO) to inhibit oncogenic miRNAs and miRNA replacement therapy for tumor
suppressor miRNAs are essential tools for miRNA based cancer therapeutics. In this review we describe the current
status of the role miRNAs play in ovarian cancer and focus on the possibilities of microRNA-based therapies and the
use of microRNAs as diagnostic tools.
Keywords: Ovarian cancer, miRNAs, Chemoresistance, Diagnosis, Prognosis, Therapy, miR-200, Let-7
Introduction
Epithelial ovarian cancer (referred to as ovarian cancer
in this review) is the fifth most common cancer among
women and causes more deaths than any other type of
female reproductive cancer [1]. Signs and symptoms of
ovarian cancer are frequently absent or ambiguous early
on and due to a lack of early detection strategies most
(>60%) patients are diagnosed with advanced-stage dis-
ease. The five year survival rate is less than 30% for these
advanced-stage patients and, despite advances in chemo-
therapy, survival rates have only modestly improved over
the past 40 years [1-3]. Pathologically, ovarian cancer is
a heterogeneous disease comprised of serous, mucinous,
clear cell, and endometrioid subtypes. Serous tumors are
the most common subtype. Each subtype is associated
with diverse genetic risk factors and molecular events
during oncogenesis and is characterized by distinct
mRNA expression profiles. It has been observed that
subtypes respond differently to chemotherapy. The re-
sponse rate of clear cell carcinomas (15%) is very low,
whereas the response rate for high-grade serous is 80%,
resulting in a lower 5-year survival for clear cell com-
pared with high-grade serous carcinoma in patients with
advanced stage tumors (20% versus 30%) [4,5].
The standard treatment for advanced ovarian cancer is
surgical tumor debulking (removal of all tumor and me-
tastasis that can be macroscopically detected in the en-
tire abdomen region), followed by platinum-based
* Correspondence: subhash.chauhan@sanfordhealth.org
1
Cancer Biology Research Center, Sanford Research/USD, 2301 East 60th
Street North, Sioux Falls SD 57104, USA
2
Department of Obstetrics and Gynecology, Sanford School of Medicine, The
University of South Dakota, 2301 East 60th Street North, Sioux Falls SD 57105,
USA
Full list of author information is available at the end of the article
© 2012 Zaman et al.; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative
Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and
reproduction in any medium, provided the original work is properly cited.
Zaman et al. Journal of Ovarian Research 2012, 5:44
http://www.ovarianresearch.com/content/5/1/44
chemotherapy [6]. Neoadjuvant therapy, the use of
chemotherapy or radiation prior to surgery, may be an
option for patients with stage IIIC or IV ovarian cancer
which typically presents with a large tumor burden and
extensive metastases. For these patients optimal debulk-
ing may be difficult to achieve, making neoadjuvant ther-
apy an important option [7]; however, there is currently
an ongoing debate to clearly identify which patients
would benefit from neoadjuvant therapy [8,9].
Chemotherapy in ovarian cancer includes platinum-
based drugs, cisplatin or carboplatin coupled with pacli-
taxel. After first-line treatment with carboplatin and
paclitaxel, most patients eventually relapse with a me-
dian progression-free survival of 18 months. Recurrent
ovarian cancer initially responds to additional chemo-
therapy; however, the progression-free interval becomes
shorter after each cycle as chemo-resistance increases
until the disease becomes incurable [10]. A number of
molecular mechanisms have been characterized to ex-
plain the development of resistance to chemotherapy,
such as increased DNA repair activity and defective
DNA damage response [11], increased anti-apoptotic
regulator activity [12,13], growth factor receptor deregu-
lation [14] [15] and post-translational modification or
aberrant expression of β-tubulin and other microtubule
regulatory proteins [16].
The recently discovered microRNAs (miRNAs) consti-
tute a novel regulatory layer of gene expression and have
been implicated in the etiology of various kinds of
human cancers. miRNAs are small (~22bp) endogenous
non-coding RNAs and are frequently dysregulated in
cancer. Their role is to modulate gene expression mainly
by base-pairing to the 3-UTR (untranslated region) of
the target mRNA, eventually causing either translational
repression, mRNA cleavage, or destabilization [17]. In
ovarian carcinoma the expression of various miRNAs
has been found to be dysregulated [18]. Recent reports
support a role for miRNAs in the initiation and progres-
sion of ovarian carcinoma [19-21] by promoting the ex-
pression of proto-oncogenes or by inhibiting the
expression of tumor suppressor genes. In this review we
focus on the current status of the role miRNAs play in
Figure 1 Oncogenic and tumor suppressor miRNAs in ovarian carcinoma. Based on their function miRNAs can be used for diagnostics and
therapeutics. Certain miRNAs such as miR-200 family, let-7 family, miR-21, miR-214 and miR-100 have strong diagnostic/prognostic potential in
ovarian cancer. Use of antagonists for oncogenic microRNAs and microRNA replacement therapy for tumor suppressor miRNAs are important
tools in miRNA based cancer treatment. EMT-Epithelial to Mesenchymal Transition; NM-Nuclear membrane; PM-Plasma membrane.
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 2 of 11
http://www.ovarianresearch.com/content/5/1/44
ovarian cancer (Figure-1 and Table-1). We also describe
their diagnostic / prognostic and therapeutic potential.
miRNAs
miRNAs were first discovered in 1993 by Lee, Feinbaum
and Ambros in the nematode C. elegans [22] and are
now known to be present and highly conserved among a
wide range of species [23]. Mature miRNAs are derived
from precursors called pri-miRNAs, composed of hun-
dreds or thousands of nucleotides [17,24,25]. miRNAs
precursor sequences are located in different parts of nu-
clear DNA and may constitute mono- or policistrone
transcriptional units. Pri-miRNAs are transcribed mainly
by polymerase RNA II. Subsequently, they are cleaved by
the endonuclease Drosha and cofactor DGCR8 into a
structure known as precursor miRNA or pre-miRNA.
Pre-miRNAs, ~60 nucleotide stem-loop molecules, are
transported from the nucleus to the cytoplasm by Expor-
tin 5 and protein Ran-GTP and further processed by the
Dicer enzyme into a ~22 nucleotide double-stranded
microRNA [26]. The double-stranded miRNA assembles
into a ribonucleoprotein complex which is known as the
RNA induced silencing complex (RISC) [27]. The RISC
induces unwinding of the double-stranded molecule into
single stranded miRNA, concomitantly degrading the
complementary strand. In animals the miRNARISC
binds to 3untranslated region (3UTR) of mRNA, does
not require perfect complementarity, and induces inhib-
ition of translation at the initiation or elongation phase
[27]. The mode of inhibition may depend, in part, on the
level of complementarity of the miRNAs where perfect
or near perfect complementarity favors degradation. The
mechanism of 3UTR mRNA target regulation is com-
plex. Nevertheless, recent studies suggest that it is a two
step process in which inhibition of translation is done
first, followed by mRNA decay due to deadenylation of
the mRNA [28]. The seed sequence is essential for the
binding of the miRNA to the mRNA. The seed sequence
is a conserved heptamerical sequence which is mostly
situated at positions 27 from the 5end of the miRNA,
although other factors are also important [17,29]. Gu
et. al. [30] have suggested that miRNA target sites can
also be found in the 5UTR or even in the coding re-
gion of the mRNA. By binding to 5UTR sequences
miRNAs can also activate translation. Thus, inhibition
of posttranscriptional mRNA processing is not the only
way of regulating miRNA-dependent gene expression
[31]. Moreover, as miRNAs do not require perfect com-
plementarity for functional interactions with mRNA tar-
gets, a single miRNA can regulate multiple targets and
conversely, multiple miRNAs are known to regulate in-
dividual mRNAs [32].
miRNAs in cancer
miRNAs are involved in crucial biological processes, in-
cluding development, differentiation, apoptosis and pro-
liferation [33]. This is done through imperfect pairing
with target messenger RNAs (mRNAs) of protein-coding
Table 1 miRNAs: functions and targets in ovarian cancer
microRNAs Function Targets Reference
miR-200 family Tumor suppressor ZEB1,ZEB2,βtubulin III [31,42,45,47,48,51-59,111,112]
(Loss of EMT)
Let-7 family Tumor suppressor KRAS,HRAS,C-MYC,HMGA-2,CyclinA,D1,D2,D3,CDC25,CDK6 [47,68-81,110,113,116,117]
(Chemosensitization)
miR-34a/b/c Tumor suppressor c-myc,CDK6,Notch-1,MET,E2f3,Bcl2,cyclinD1 [82,119]
(Reduced invasion/migration/proliferation)
miR-100 Tumor suppressor mTOR, PLK-1 [83-85]
(Increased sensitivity to rapamycin analogs)
miR-31 Tumor suppressor E2F2, STK40 [86]
(Increased apoptosis)
miR-214/199a* Oncogenic PTEN [51,87-91,93,112]
(Chemoresistance)
miR-376c Oncogenic ALK7 [21,98]
(Chemoresistance)
miR-93 Oncogenic PTEN [100-102]
(Chemoresistance)
miR-21 Oncogenic PTEN [47,51,104-107,112]
(Chemoresistance)
miRNAs in ovarian carcinoma can either be oncogenic or tumor suppressor. Tumor suppressor miRNAs suppress oncogenes resulting in either loss of EMT
(Epithelial Mesenchymal Transition), chemosensitization or tumor suppression. Whereas, oncogenic miRNAs target tumor suppressor genes leading to chemo
resistance and reduced survival.
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 3 of 11
http://www.ovarianresearch.com/content/5/1/44
genes and the transcriptional or post-transcriptional
regulation of their expression [26]. The first reported
link between miRNAs and cancer regarded chronic
lymphocytic leukemia, wherein miR-15 and miR-16 were
found to be deleted or down-regulated in a majority of
tumors [34]. Since then, changes in the expression level
of miRNAs have subsequently been detected in many
types of human tumors [35]. miRNAs have various roles
in oncogenesis as they can function either as tumor sup-
pressors (e.g., miR-15a and miR-16-1) or oncogenes (e.g.,
miR-155 or members of the miR-1792 cluster). Recent
studies on the abnormal expression of miRNAs in can-
cer have described the following reasons for differential
expression: chromosomal rearrangements [36-38], gen-
omic copy number change [39,40], epigenetic modifica-
tions [41,42], defects in miRNA biogenesis pathway [43],
and regulation by transcription factors [44].
miRNAs in ovarian cancer
One of the seminal studies done on epithelial ovarian
cancer (EOC) and miRNA was done by Zhang et. al. in
2008 [45]. In this study the authors utilized an integra-
tive genomic approach to study miRNA deregulation in
human epithelial ovarian cancer. They compared mature
miRNA expression profiles in 18 ovarian cancer cell
lines and 4 immortalized, non neoplastic cell lines
derived from normal ovarian surface epithelium. Thirty
five miRNAs were found to be differentially expressed
between the two groups of cells. Out of these 35, only 4
were up-regulated, whereas the rest were mostly down-
regulated in cancer cells as compared to immortalized
non-neoplastic cell lines. The 4 up-regulated miRNAs
were miR-26b, miR-182, miR-103 and miR-26a. The list
of down-regulated miRNAs included prominent tumor
suppressors such as let-7d and miR-127. This showed
that miRNA expression profiles can distinguish malig-
nant from nonmalignant ovarian surface epithelium.
Zhang et. al. went on to analyze 106 primary human
ovarian cancer specimens of various stages and grades
using miRNA microarrays. They found that all tumor
suppressor miRNA alterations were related to down-
regulation in late stage tumors, which included miR-15a,
miR-34a and miR-34b. The authors also observed that
DNA copy number loss and epigenetic silencing are
mainly responsible for down-regulation of the miRNAs.
In the case of over-expressed miRNAs (oncogenic miR-
NAs such as miR-182) the chromosomal regions were
found to be amplified in a significant number of cancer
samples. Additionally, epigenetic alterations reduced the
expression of 16 out of 44 miRNAs which were down-
regulated in late-stage ovarian cancer as the expression
of these miRNAs, e.g. miR-34b, was restored by DNA
demethylation or histone deacetylase inhibiting agents.
In addition several studies have compared the expres-
sion profile of miRNAs in a large number of clinical
ovarian cancer samples to normal ovarian tissues, ovar-
ian epithelial cell lines or fallopian tubes [42,46-51]. The
following sections provide information regarding miR-
NAs that are suggested to be involved in the pathobiol-
ogy of ovarian cancer (Tables 1 and 2).
miRNA-200 family
The miR-200 family contains miR-200a, miR-200b, miR-
200c, miR-141 and miR-429 which are arranged in 2
clusters in the human genome. miR-200a, miR-200b and
miR-429 are located on chromosome 1, while miR-200c
and miR-141 are on chromosome 12 [52]. Iorio et. al.
[42] have shown that the miR-200 family is among the
most significantly over-expressed miRNAs in epithelial
ovarian cancer. The expression of miR-200a and miR-
200c was found to be up-regulated in three types of
ovarian cancer: serous, endometrioid and clear cell.
However, miR-200b and miR-141 are up-regulated in
endometrioid and serous subtypes. The role of the miR-
200 family in ovarian carcinoma is complicated. While
miR-200 family members are believed to be metastasis
suppressants, the majority of studies done on the family
relate to over expression in ovarian cancer. However,
some studies report that miR-200 family members are
either down regulated [48] or even unchanged [45].
These differing results may occur because of the use of
different normal controls or the inclusion of ovarian
stromal cells which lack miR200 expression.
Further complicating the potential roles of the miR-
200 family members, recent studies have implicated the
miR-200 family with the regulation of the epithelial to
Table 2 miRNA profile of subtypes of ovarian cancer
Type of ovarian
cancer
Up-regulated miRNA Down-regulated miRNA References
Serous miR-200a, miR-200b, miR-200c, miR-141, miR-93, miR-21,
miR-519a, miR-214
let7-b, miR-99a, miR-125b, miR-22, miR-
31, miR-34a/b/c
[42,47,51,86,102,106,108,109]
Clear Cell miR-519a, miR-182, miR-30a,miR-21, miR-200a, miR-200c miR-100, miR-22, miR-34a/b/c, miR-214 [42,51,83,109]
Mucinous miR-153, miR-485-5p [109]
Endometrioid miR-200a, miR-200b, miR-200c, miR-141 [42]
Most of the miRNA profiling has been done on the serous and clear cell ovarian carcinoma. miR-200 family stands out as the up-regulated miRNA in most types
of ovarian cancer. miR-100 plays a specific role in clear cell ovarian carcinoma.
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 4 of 11
http://www.ovarianresearch.com/content/5/1/44
mesenchymal transition (EMT). EMT is a process where
epithelial tumor cells are stimulated by extracellular
cytokines, e.g. TGFβ, or intracellular molecules such as
oncogenic Ras, to change their epithelial characteristics
into a mesenchymal phenotype with increased migratory
and invasive capabilities. EMT is induced by a group of
transcriptional repressors, such as Snail, Slug, TWIST,
Id2, ZEB1 and ZEB2. The protein levels of these re-
pressors increase during EMT, resulting in the down-
regulation of genes such as E-cadherin which are
responsible for the epithelial identity of the cells [53].
The E-cadherin molecules mediate cell-cell adhesion.
Park et. al. [54] have shown a positive correlation in the
expression of E-cadherin with the expression of miR-
200c in ovarian cancer tissues. The miR-200 family
members have also been shown to suppress the expres-
sion of ZEB1 and ZEB2, thereby suppressing EMT.
Over expression of miR-200 a/b/c and/or miR-141
down regulates ZEB1/2 levels, and leads to higher
levels of E-cadherin and an epithelial phenotype. On
the contrary, ZEB1/2 can inhibit the expression of
miR-200 family members by binding to the promoter
of both miR-200 clusters thereby blocking transcrip-
tion. The mechanistic explanation of the above process
can be summarized as follows: activation of a trigger
such as TGF-βor PDGF-D [52], leads to increased
levels of ZEB1/2, decreased expression of miR-200 and
the induction of EMT [31,54-57]. The miR-200 family
might be down regulated when cancer cells acquire in-
vasive properties and then become up-regulated when
the cells undergo mesenchymal to epithelial transition
during the process of re-epithelialization, which is evi-
dent from the positive correlation of miR-200 and E-
cadherin expressions [58].
In a recent study, Leskela et. al. [59] demonstrated the
role of the miR-200 family members in controlling β-
tubulin III expression and its association with paclitaxel-
based treatment response and progression-free survival
in ovarian cancer patients. Previous studies demon-
strated that high tumoral β-tubulin III expression has
been associated with decreased survival with non-small
cell lung cancer [60,61], breast [62], head and neck [63]
and ovarian cancer [64]. Moreover, a number of studies
have also shown that high expression of β-tubulin III is
associated with worse treatment response in ovarian
cancer [65-67]. Leskela et. al. found that tumors with
high levels of β-tubulin III protein have significantly
decreased miR-200 expression. They observed the stron-
gest associations with miR-141, miR-429 and miR-200c.
miR-200c expression was statistically significantly asso-
ciated with response to treatment as patients who did
not achieve a complete clinical response had lower levels
of miR-200c as compared to those showing a complete
response. Low expression of miR-200c was also
associated with recurrence of ovarian cancer. Moreover,
miR-429 expression was found to be significantly asso-
ciated with recurrence-free survival and overall survival
of the patients [59].
Let-7 family
The let-7 (lethal-7) family in humans consists of 13 miR-
NAs located on nine different chromosomes [68,69]. In
multiple human cancers expression of the let-7 family is
significantly reduced. Low let-7 expression has been
found to be associated with poor survival of cancer
patients [70,71]. Let-7 suppresses multiple ovarian can-
cer oncogenes, which includes KRAS,HRAS[72], c-MYC
[72] and HMGA-2 [73]. Moreover, it also inhibits cell
cycle regulators such as CDC25, CDK6 as well as Cyclin
A, D1, D2 and D3 [74,75]. The mechanism of down-
regulation for let-7 miRNAs is through copy-number
alterations [76]. The genomic locus containing let-7a-3/
let-7b was deleted in 44% of ovarian cancer samples
studied. Restoration of let-7b expression significantly
reduced ovarian tumor growth both in vitro and in vivo.
Additionally, recent studies have shown a correlation
between loss of let-7 and resistance to either chemother-
apeutic drugs or radiation [71,77-79]. Using a drug re-
sistant ovarian cancer cell line, Boyerinas et. al. [80]
demonstrated that drug sensitivity to taxanes is
increased upon over expression of let-7g as it inhibits
IMP-1, an RNA binding protein which stabilizes the
mRNA of a number of target genes, including, MDR1
(multidrug resistance-1). MDR1 is a member of the ad-
enosine triphosphate binding cassette transporters (ABC
transporter family) which pump drugs across the cell
membrane to the extracellular space. Therefore, the ex-
pression of let-7g resulted in a decrease in MDR1 and
sensitized the cells to Taxane treatment.
On the other hand, Lu et. al. [81] observed that ovar-
ian cancer patients responding to a regimen of platinum
and paclitaxel had significantly lower let-7a expression
than those who did not respond to treatment. Moreover,
survival data indicated that patients with high let-7a sur-
vived better when they were treated with platinum only
(no paclitaxel) as compared to those having low expres-
sion of let-7a. The authors conclude that miRNA-let-7a
expression can be a potential marker for selection of
chemotherapeutic agents in ovarian cancer treatment.
miRNA-34 a/b/c
Quantitative-RT PCR and in-situ hybridization in a
panel of 83 human ovarian cancer samples showed a sig-
nificant decrease in miR-34a/b/c expression. The de-
crease was also correlated with the p53 status as p53
regulates the expression of miR-34 family members by
promoter methylation and copy number alterations [82].
Over-expression of miR-34 family members reduced
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 5 of 11
http://www.ovarianresearch.com/content/5/1/44
migration, invasion and cellular proliferation in ovarian
cancer cell lines, providing evidence that the loss of
miR-34 family members may be involved in the patho-
biology of ovarian cancer.
miRNA-100
miR-100 is a tumor suppressor which has been found to
be down-regulated in most of the ovarian cancer cell
lines, especially clear cell ovarian carcinoma cell lines
and ovarian cancer tissues [83,84]. miR-100 represses
mTOR (mammalian target of rapamycin) signaling and
increases sensitivity to the cancer drug everolimus (rapa-
mycin analog RAD001) in cell lines derived from clear
cell carcinomas. mTOR is a serine/threonine kinase and
is a downstream effector of the Akt signaling pathway.
mTOR has also been shown to be a possible therapeutic
target in both cisplatin-sensitive and cisplatin-resistant
clear cell ovarian carcinoma [85]. Low miR-100 expres-
sion was associated with shorter overall patient survival
and advanced stage ovarian cancer. Moreover, its expres-
sion has been shown to be an independent predictor of
overall survival in ovarian cancer patients. miR-100 also
inhibits the expression of the proto-oncogene PLK1
(Polo-like kinase-1) in ovarian cancer [84].
miRNA-31
miR-31 is under-expressed in both serous ovarian cancer
cell lines and tissues [86]. miRNA-31 inhibits the expres-
sion of cell cycle regulators such as E2F2 and STK40, a
repressor of p53 mediated transcription, and acts as a
tumor suppressor in ovarian cancer. Over expression of
miR-31 in ovarian cancer cell lines having non func-
tional p53 pathways lead to decreased proliferation and
increased caspase-mediated apoptosis, whereas, over ex-
pression of miR-31 had no effect on ovarian cancer cells
having wild-type p53 [86]. Thus, miRNA-31 might have
therapeutic roles in the case of cancers having p53
mutations.
miRNA-214/199a*
Up-regulation of miR-214 has been detected in various
human malignancies, including pancreatic, prostate, gas-
tric, breast and ovarian cancers as well as malignant
melanoma [51,87-90]. miR-214 has extensive roles in
chemo-resistance, tumor progression and metastasis
[51,87,88,91]. Yang et. al. [51] have shown that miR-214
induces cell survival and cisplatin resistance by targeting
PTEN. miRNA microarrays show the aberrant regulation
of 36 miRNAs between normal ovarian cells and epithe-
lial ovarian tumors [51]. miR-199a*, miR-214, miR-200a
and miR-100 were most highly differentially expressed.
miR-199a* and miR-214 were found to be up-regulated
in 53 and 56% of the tumor tissues, respectively. miR-
214 knockdown was found to abrogate cisplatin
resistance in cisplatin-resistant cell line A2780CP,
whereas exogenous expression of miR-214 renders
cisplatin-sensitive cell line A2780S and OV119 cells re-
sistant to cisplatin induced apoptosis. miR-214 activates
the Akt pathway by targeting PTEN, which normally
negatively regulates Akt. Constitutive activation of Akt
leads to chemo-resistance in different types of tumors
including ovarian cancer [92]. Thus, miR-214 possibly
plays an important role in cisplatin resistance by target-
ing the PTEN/Akt pathway. miR-199a and miR-214 have
been implicated in the process of differentiation of ovar-
ian cancer stem cells (CSCs) into mature ovarian cancer
cells [93]. Twist 1, a transcription factor belonging to
basic helix-loop-helix proteins has been shown to regu-
late the expression of both miR-199a and miR-214
which are part of the human Dnm3os gene. Twist 1 is
involved in the differentiation of multiple cell lineages,
including muscle, cartilage and osteogenic cells [94-97].
Twist 1 levels increase during the differentiation process
leading to an increase in miR-199a and miR-214, a de-
crease in IKKβexpression (target of miR-199a), and a
decrease in PTEN expression (target of miR-214). This
eventually results in an increase in the pAkt activity
leading to the process of differentiation.
miRNA-376c
miRNA-376c was earlier known as miR-368 and was
found to be over expressed in a subset of acute myeloid
leukemia [98]. Ye et. al. have shown that miR-376c pro-
motes cell proliferation, survival and spheroid formation
in ovarian cancer cells [21]. This is done by suppressing
activin receptor-like kinase 7 (ALK7) and its ligand
Nodal, which together are able to induce apoptosis in
human epithelial ovarian cancer cells. A previous study
had demonstrated that the Nodal-ALK7 pathway might
be involved in chemosensitivity [99]. miR-376c over ex-
pression significantly reduced the effect of cisplatin.
Moreover, miR-376c and siRNA inhibitors of Nodal
and ALK7 also blocked the effect of carboplatin.
Chemosensitive and chemoresistant ovarian tumors
showed a differential expression of ALK7 and miR-
376c. Immuno-histochemical staining was used to stain
tumors with ALK7 and miR-376c was detected using
real-time PCR, in patients showing a complete response
(CR) and those who had an incomplete response (IR).
Patients showing CR showed significant ALK7 staining,
whereas the staining intensity was very weak in patients
with IR. Additionally, the miR-376c expression level
was inversely related to ALK7 in both cases [21].
miRNA-93
miRNA-93 is part of the miR-106b-25 cluster [100]. It
has been shown to promote tumor growth and angio-
genesis by targeting integrin-β8 [101]. In ovarian cancer
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 6 of 11
http://www.ovarianresearch.com/content/5/1/44
it is up-regulated in cisplatin-resistant ovarian cancer
cells [102]. It regulates cisplatin chemosensitivity in cis-
platin resistant ovarian cancer cells OVCAR3 and
SKOV3 by targeting the phosphatase PTEN. Over-
expression of miR-93 in both these cells increased the
ratio of phosphorylated Akt/over the total Akt (pAkt/
total Akt). Phospho rylated-Akt has been shown to play
an important role in multiple drug resistance including
cisplatin [92,103].
miRNA-21
miR-21 is aberrantly expressed and functions as an
oncogenic miRNA in many tumors including ovarian
cancer [51,104,105]. In ovarian cancer cells it promotes
cell proliferation, invasion and migration through target-
ing PTEN [106]. miR-21 also has a role in resistance to
hypoxic conditions which inhibit tumor growth [107].
Protein kinase Akt2 induces miR-21 expression under
oxygen deprivation leading to suppression of tumor sup-
pressor proteins PTEN, PDCD4 and Sprouty 1 (targets
of miR-21). This results in resistance to hypoxia [107].
miRNA expression profiles of subtypes of ovarian cancer
In addition to the miRNAs discussed above, a number of
studies have profiled the miRNA expression of specific
subtypes of ovarian cancer. Nam et. al., used a custo-
mized miRNA microarray of 314 human miRNAs to
analyze the miRNA expression profiles of serous ovarian
cancer tissues as compared to normal ovarian tissues
[47]. They found differential expression of 23 miRNAs.
miR-21 was most frequently up-regulated and miR-125b
was most frequently down- regulated (Table-2). North-
ern blot analysis confirmed the up-regulation of miR-
200c, miR-93 and miR-141 and the down-regulation of
let-7b, miR-99a and miR-125b. In a separate study, Li et.
al. have shown a negative correlation between miR-22
expression and the metastatic potential in serous ovarian
cancer cell lines [108]. Additionally, miR-519a was found
to be significantly up-regulated in serous and clear cell
carcinomas as compared to the mucinous subtype in tis-
sue samples [109]. Higher expression of miR-519a in late
stage serous carcinoma showed positive correlation with
poor progression-free survival. miR-153 and miR-485-5p
were found to be up-regulated in mucinous ovarian car-
cinoma. The down-regulation of miR-153 and miR-485-
5p showed significant correlation with advanced clinical
stage FIGO (International Federation of Gynecology and
Obstetrics) grade 3 and miR-519a was found to be high
in clinical stages III and IV (advanced clinical stages) as
compared to stages I and II (early clinical stages) [109].
As mentioned above miR-100 has been shown to be
down-regulated in clear cell ovarian carcinoma cell lines
and its over-expression in them inhibited mTOR signal-
ing and enhanced sensitivity to rapamycin analog
RAD001 (everolimus) [83] (Table-2). In the same study
the authors show the down-regulation of miR-22 and
the up-regulation of miR-182 and miR-30a in clear cell
ovarian carcinoma cell lines. Over-expression of miR-22
and knockdown of miR-182 could alter the global gene
expression pattern of clear cell ovarian cell lines towards
a normal state [83].
miRNAs in ovarian cancer diagnostics / prognostics
Previous studies in ovarian carcinoma have shown that
miRNAs can be used in its diagnosis as well as progno-
sis. Lu et. al. [110] demonstrated that patients having
low let-7a-3 methylation had overall worse survival than
those with high methylation. The miRNA-200 family
plays an important role in ovarian cancer and it has been
shown that the miR-200 family cluster, which includes
miR-200a, miR-200b and miR-429, can predict poor sur-
vival when they are expressed at low levels [111]. Yang
et. al. [51] showed that miR-214, miR-199* and miR-
200a were associated with high-grade and late stage
tumors. In an interesting ovarian cancer study [112] the
authors profiled miRNA signatures from tumor-derived
exosomes. Levels of 8 miRNAs, which were previously
shown to have diagnostic potential (miR-21, 141, 200a,
200c, 200b, 203, 205 and miR-214), were compared in
exosomes isolated from serum specimens of women
with benign disease and various stages of cancer. The 8
miRNAs had similar expressions between cellular and
exosomal miRNAs, with no detection of exosomal miR-
NAs in control samples. The profile of exosomal miR-
NAs from ovarian cancer patients was distinctly
different from patients with benign disease. HMGA2/let-
7 ratio has also been used for prognostic studies [113].
High-mobility group AT-hook 2 (HMGA2), an early em-
bryonic gene is a target of miRNAs let-7a, let-7c and
let-7g. Higher HMGA2/let-7 ratio exhibited decreased
5-year progression-free survival (<10%) as compared to
a lower ratio (~40%).
Potential role of miRNAs in ovarian cancer therapeutics
miRNA therapeutics in ovarian cancer can take differ-
ent forms. Oncogenic miRNAs can be inhibited by
using antisense oligonucleotides, antagomirs, sponges
or locked nucleic acid (LNA) constructs [114]. Cancer
cells have dysregulation in several miRNAs at the
same time and targeting a single miRNA is not suffi-
cient for treatment. Multiple-target anti-miRNA anti-
sense oligodeoxyribonucleotide-MTG-AMO (Multiple
target-O-modified antagomirs) are used to inhibit mul-
tiple miRNAs at the same time [115]. The expression
of tumor suppressor miRNAs can be restored by
miRNA replacement therapy. Several miRNAs have
been used for this purpose. Use of let-7 miRNA
mimetics is a potential tool as intra-tumoral delivery
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 7 of 11
http://www.ovarianresearch.com/content/5/1/44
of let-7b has been shown to decrease the tumor bur-
den in lung tumors [116,117]. The miR-143-145 clus-
ter has been shown to be frequently deleted in
cancer. miR-143 and 145, delivered intravenously to
subcutaneous and orthotopic xenografts downregulated
the oncogenes RREB1 and KRAS [118]. One of the
more beneficial miRNA therapies is miR-34 replace-
ment therapy. P53 protein is known to enhance miR-
34 expression and it is mutated in many cancers
[119]. The intra-tumoral delivery of miR-34 mimics
impaired tumorigenesis on a xenograft model of non-
small cell lung cancer, and systemic delivery of miR-
34 reduced tumor growth of KrasLSL-G12D
+
mice
[117,120]. Certain small molecule compounds like
enoxacin have been shown to restore downregulated
miRNAs to a normal miRNA level or expression pat-
tern without affecting normal cells and with no tox-
icity in in vivo models [121,122].
miRNAs can also be used to sensitize tumors to
chemotherapy. The efflux of anticancer drugs by ABC
transporters is one of the main reasons resistance to
chemotherapy drugs develops. miR-9 has been shown to
negatively regulate SOX2 which induces the expression
of ABC transporters ABCC3 and ABCC6 [123]. Resist-
ance to tamoxifen is restored by the over expression of
miR-15 and miR-16. miR-15 and 16 suppress the anti-
apoptotic molecule BCL-2 and sensitize the cells to tam-
oxifen [124]. Similarly, use of antagomirs against miR-21
was found to sensitize cultured cells to the chemothera-
peutic agent 5-Fluorouracil (5-FU) [125].
One of the greatest challenges in RNAi therapy con-
tinues to be the delivery method of the therapeutic
siRNA or miRNA to the target cells. Future focus should
be aimed at addressing these issues by engineering an ef-
ficient delivery system by use of radiolabeled, tumor spe-
cific antibody conjugated nanoformulations to deliver
miRNA to the ovarian tumor site. It is very important
to formulate an effective delivery method, such as
nanotechnology-based delivery approach for microRNA
for therapy with the help of strategic image-guided
systemic delivery to the tumor. This will be a major
contribution in the field of cancer therapeutics and
will help overcome challenges in miRNA delivery. The
goal of this novel therapeutic approach is to effectively
encourage reprogramming of miRNA networks in can-
cer cells which may lead to a clinically translatable
miRNA-based therapy to benefit ovarian cancer
patients.
Conclusions
In spite of all the above advances there is still a long way
to go to understand and apply miRNA therapeutics in
cancer and ovarian cancer in particular. Identification of
unique patterns of deregulated miRNA expression in
ovarian cancer provides valuable information that may:
serve as molecular biomarkers for tumor diagnosis; iden-
tify low and high risk populations of patients, disease
prognosis and prevention of cancer, and predict thera-
peutic responses. One of the areas of improvement in
miRNA therapy has been to reduce or eliminate off-
target or non-specific effects. This regulatory network is
complex because of the fact that a single miRNA can
have multiple targets and several miRNAs can have a
single target. Therefore, careful designing of therapeutic
strategies is needed to overcome these technical issues.
Moreover, the identification of new strategies is required
to enhance the potency and stability of therapeutic
vectors and the specificity of their delivery to tissues.
We hope that with increased understanding of the role
of miRNAs in cancer development and by designing
more efficient miRNA-modulating molecules, miRNA
mediated cancer therapy will give a new impetus to
the cure for cancer including ovarian cancer.
Abbreviations
miRNA: MicroRNA; EMT: Epithelial to mesenchymal transition;
RISC: RNAinduced silencing complex; UTR: Untranslated region;
EOC: Epithelial ovarian cancer/ovarian cancer.
Competing interests
The authors declare that there are no financial and non financial competing
interests.
Authorscontributions
MSZ designed and drafted the manuscript; MJ, DMM and SK were involved
in the critical revision of the manuscript; SCC gave final approval of the
version to be published. All authors read and approved the final manuscript.
Acknowledgements
The authors thank Cathy Christopherson for editorial assistance and
Dr. Mohammed Sikander for preparing the figure. This work was partially
supported by grants to SCC from Governors Cancer 2010, the National
Institutes of Health Research Project Grant Program (RO1) (CA142736), and
(UO1) (CA162106A) and the Centers of Biomedical Research Excellence
(P20 RR024219).
Author details
1
Cancer Biology Research Center, Sanford Research/USD, 2301 East 60th
Street North, Sioux Falls SD 57104, USA.
2
Department of Obstetrics and
Gynecology, Sanford School of Medicine, The University of South Dakota,
2301 East 60th Street North, Sioux Falls SD 57105, USA.
3
Basic Biomedical
Science Division, Sanford School of Medicine, The University of South
Dakota, 2301 East 60th Street North, Sioux Falls SD 57105, USA.
Received: 20 October 2012 Accepted: 6 December 2012
Published: 13 December 2012
References
1. Siegel R, Naishadham D, Jemal A: Cancer statistics, 2012. CA Cancer J Clin
2012, 62(1):1029.
2. Wright JD, Shah M, Mathew L, Burke WM, Culhane J, Goldman N, Schiff PB,
Herzog TJ: Fertility preservation in young women with epithelial ovarian
cancer. Cancer 2009, 115(18):41184126.
3. Fung-Kee-Fung M, Oliver T, Elit L, Oza A, Hirte HW, Bryson P: Optimal
chemotherapy treatment for women with recurrent ovarian cancer.
Curr Oncol 2007, 14(5):195208.
4. Takano M, Kikuchi Y, Yaegashi N, Kuzuya K, Ueki M, Tsuda H, Suzuki M,
Kigawa J, Takeuchi S, Moriya T, et al:Clear cell carcinoma of the ovary: a
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 8 of 11
http://www.ovarianresearch.com/content/5/1/44
retrospective multicentre experience of 254 patients with complete
surgical staging. Br J Cancer 2006, 94(10):13691374.
5. du Bois A, Luck HJ, Meier W, Adams HP, Mobus V, Costa S, Bauknecht T,
Richter B, Warm M, Schroder W, et al:A randomized clinical trial of
cisplatin/paclitaxel versus carboplatin/paclitaxel as first-line treatment of
ovarian cancer. J Natl Cancer Inst 2003, 95(17):13201329.
6. Shih Ie M, Kurman RJ: Ovarian tumorigenesis: a proposed model based
on morphological and molecular genetic analysis. Am J Pathol 2004,
164(5):15111518.
7. Morgan RJ, Alvarez RD, Armstrong DK, Burger RA, Castells M, Chen LM,
Copeland L, Crispens MA, Gershenson D, Gray H, et al:Ovarian Cancer,
Version 3.2012. J Natl Compr Canc Netw 2012, 10(11):13391349.
8. Vergote I, du Bois A, Amant F, Heitz F, Leunen K, Harter P: Neoadjuvant
chemotherapy in advanced ovarian cancer: On what do we agree and
disagree. Gynecol Oncol 2012, Sep 21 (Epub ahead of print).
9. Gonzalez-Martin A, Chiva L: Emerging Concerns When Evidence-Based
Medicine Is Translated into Real Life: The Case of Neoadjuvant
Chemotherapy in Ovarian Cancer. Curr Oncol Rep 2012, Oct 9
(epub ahead of print).
10. Cannistra SA: Cancer of the ovary. N Engl J Med 2004, 351(24):25192529.
11. Rabik CA, Dolan ME: Molecular mechanisms of resistance and toxicity
associated with platinating agents. Cancer Treat Rev 2007, 33(1):923.
12. Fraser M, Bai T, Tsang BK: Akt promotes cisplatin resistance in human
ovarian cancer cells through inhibition of p53 phosphorylation and
nuclear function. Int J Cancer 2008, 122(3):534546.
13. Behbakht K, Qamar L, Aldridge CS, Coletta RD, Davidson SA, Thorburn A,
Ford HL: Six1 overexpression in ovarian carcinoma causes resistance to
TRAIL-mediated apoptosis and is associated with poor survival. Cancer
Res 2007, 67(7):30363042.
14. Duan Z, Foster R, Bell DA, Mahoney J, Wolak K, Vaidya A, Hampel C, Lee H,
Seiden MV: Signal transducers and activators of transcription 3 pathway
activation in drug-resistant ovarian cancer. Clin Cancer Res 2006,
12(17):50555063.
15. Gan Y, Wientjes MG, Au JL: Expression of basic fibroblast growth factor
correlates with resistance to paclitaxel in human patient tumors.
Pharm Res 2006, 23(6):13241331.
16. Ferlini C, Raspaglio G, Cicchillitti L, Mozzetti S, Prislei S, Bartollino S, Scambia
G: Looking at drug resistance mechanisms for microtubule interacting
drugs: does TUBB3 work? Curr Cancer Drug Targets 2007, 7(8):704712.
17. Bartel DP: MicroRNAs: genomics, biogenesis, mechanism, and function.
Cell 2004, 116(2):281297.
18. Bartels CL, Tsongalis GJ: MicroRNAs: novel biomarkers for human cancer.
Clin Chem 2009, 55(4):623631.
19. Sorrentino A, Liu CG, Addario A, Peschle C, Scambia G, Ferlini C: Role of
microRNAs in drug-resistant ovarian cancer cells. Gynecol Oncol 2008,
111(3):478486.
20. Marchini S, Fruscio R, Clivio L, Beltrame L, Porcu L, Nerini IF, Cavalieri D,
Chiorino G, Cattoretti G, Mangioni C, et al:Resistance to platinum-based
chemotherapy is associated with epithelial to mesenchymal transition in
epithelial ovarian cancer. Eur J Cancer 2012, Aug 13, (Epub ahead of print).
21. Ye G, Fu G, Cui S, Zhao S, Bernaudo S, Bai Y, Ding Y, Zhang Y, Yang BB,
Peng C: MicroRNA 376c enhances ovarian cancer cell survival by
targeting activin receptor-like kinase 7: implications for chemoresistance.
J Cell Sci 2011, 124(Pt 3):359368.
22. Lee RC, Feinbaum RL, Ambros V: The C. elegans heterochronic gene lin-4
encodes small RNAs with antisense complementarity to lin-14. Cell 1993,
75(5):843854.
23. Wheeler BM, Heimberg AM, Moy VN, Sperling EA, Holstein TW, Heber S,
Peterson KJ: The deep evolution of metazoan microRNAs. Evol Dev 2009,
11(1):5068.
24. Ambros V: The functions of animal microRNAs. Nature 2004,
431(7006):350355.
25. Cai X, Hagedorn CH, Cullen BR: Human microRNAs are processed from
capped, polyadenylated transcripts that can also function as mRNAs.
RNA 2004, 10(12):19571966.
26. Kim YK, Kim VN: Processing of intronic microRNAs. EMBO J 2007,
26(3):775783.
27. Pratt AJ, MacRae IJ: The RNA-induced silencing complex: a versatile
gene-silencing machine. J Biol Chem 2009, 284(27):1789717901.
28. Fabian MR, Mathonnet G, Sundermeier T, Mathys H, Zipprich JT, Svitkin YV,
Rivas F, Jinek M, Wohlschlegel J, Doudna JA, et al:Mammalian miRNA RISC
recruits CAF1 and PABP to affect PABP-dependent deadenylation.
Mol Cell 2009, 35(6):868880.
29. Grimson A, Farh KK, Johnston WK, Garrett-Engele P, Lim LP, Bartel DP:
MicroRNA targeting specificity in mammals: determinants beyond seed
pairing. Mol Cell 2007, 27(1):91105.
30. Gu S, Jin L, Zhang F, Sarnow P, Kay MA: Biological basis for restriction of
microRNA targets to the 3' untranslated region in mammalian mRNAs.
Nat Struct Mol Biol 2009, 16(2):144150.
31. Vasudevan S, Tong Y, Steitz JA: Cell-cycle control of microRNA-mediated
translation regulation. Cell Cycle 2008, 7(11):15451549.
32. Lewis BP, Shih IH, Jones-Rhoades MW, Bartel DP, Burge CB: Prediction of
mammalian microRNA targets. Cell 2003, 115(7):787798.
33. Flynt AS, Lai EC: Biological principles of microRNA-mediated regulation:
shared themes amid diversity. Nat Rev Genet 2008, 9(11):831842.
34. Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H,
Rattan S, Keating M, Rai K, et al:Frequent deletions and down-regulation
of micro- RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic
leukemia. Proc Natl Acad Sci U S A 2002, 99(24):1552415529.
35. Zhang W, Dahlberg JE, Tam W: MicroRNAs in tumorigenesis: a primer.
Am J Pathol 2007, 171(3):728738.
36. Calin GA, Ferracin M, Cimmino A, Di Leva G, Shimizu M, Wojcik SE, Iorio MV,
Visone R, Sever NI, Fabbri M, et al:A MicroRNA signature associated with
prognosis and progression in chronic lymphocytic leukemia. N Engl J
Med 2005, 353(17):17931801.
37. Calin GA, Croce CM: Chromosomal rearrangements and microRNAs: a
new cancer link with clinical implications. J Clin Invest 2007,
117(8):20592066.
38. Tagawa H, Seto M: A microRNA cluster as a target of genomic
amplification in malignant lymphoma. Leukemia 2005, 19(11):20132016.
39. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S,
Shimizu M, Rattan S, Bullrich F, Negrini M, et al:Human microRNA genes
are frequently located at fragile sites and genomic regions involved in
cancers. Proc Natl Acad Sci U S A 2004, 101(9):29993004.
40. Giannakakis A, Sandaltzopoulos R, Greshock J, Liang S, Huang J, Hasegawa
K, Li C, O'Brien-Jenkins A, Katsaros D, Weber BL, et al:miR-210 links hypoxia
with cell cycle regulation and is deleted in human epithelial ovarian
cancer. Cancer Biol Ther 2008, 7:255264.
41. Saito Y, Liang G, Egger G, Friedman JM, Chuang JC, Coetzee GA, Jones PA:
Specific activation of microRNA-127 with downregulation of the proto-
oncogene BCL6 by chromatin-modifying drugs in human cancer cells.
Cancer Cell 2006, 9(6):435443.
42. Iorio MV, Visone R, Di Leva G, Donati V, Petrocca F, Casalini P, Taccioli C,
Volinia S, Liu CG, Alder H, et al:MicroRNA signatures in human ovarian
cancer. Cancer Res 2007, 67(18):86998707.
43. Kumar MS, Lu J, Mercer KL, Golub TR, Jacks T: Impaired microRNA
processing enhances cellular transformation and tumorigenesis. Nat
Genet 2007, 39(5):673677.
44. He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Zender L,
Magnus J, Ridzon D, et al:A microRNA component of the p53 tumour
suppressor network. Nature 2007, 447(7148):11301134.
45. Zhang L, Volinia S, Bonome T, Calin GA, Greshock J, Yang N, Liu CG,
Giannakakis A, Alexiou P, Hasegawa K, et al:Genomic and epigenetic
alterations deregulate microRNA expression in human epithelial ovarian
cancer. Proc Natl Acad Sci U S A 2008, 105(19):70047009.
46. Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S,
Naylor TL, Barchetti A, Ward MR, et al:microRNAs exhibit high frequency
genomic alterations in human cancer. Proc Natl Acad Sci U S A 2006,
103(24):91369141.
47. Nam EJ, Yoon H, Kim SW, Kim H, Kim YT, Kim JH, Kim JW, Kim S: MicroRNA
expression profiles in serous ovarian carcinoma. Clin Cancer Res 2008,
14(9):26902695.
48. Dahiya N, Sherman-Baust CA, Wang TL, Davidson B, Shih Ie M, Zhang Y,
Becker KG, Morin PJ, Wood W: MicroRNA expression and identification
of putative miRNA targets in ovarian cancer. PLoS One 2008,
3(6):e2436.
49. Wyman SK, Parkin RK, Mitchell PS, Fritz BR, O'Briant K, Godwin AK, Urban N,
Drescher CW, Knudsen BS, Tewari M: Repertoire of microRNAs in epithelial
ovarian cancer as determined by next generation sequencing of small
RNA cDNA libraries. PLoS One 2009, 4(4):e5311.
50. Lee CH, Subramanian S, Beck AH, Espinosa I, Senz J, Zhu SX, Huntsman D,
van de Rijn M, Gilks CB: MicroRNA profiling of BRCA1/2 mutation-carrying
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 9 of 11
http://www.ovarianresearch.com/content/5/1/44
and non-mutation-carrying high-grade serous carcinomas of ovary.
PLoS One 2009, 4(10):e7314.
51. Yang H, Kong W, He L, Zhao JJ, O'Donnell JD, Wang J, Wenham RM,
Coppola D, Kruk PA, Nicosia SV, et al:MicroRNA expression profiling in
human ovarian cancer: miR-214 induces cell survival and cisplatin
resistance by targeting PTEN. Cancer Res 2008, 68(2):425433.
52. Korpal M, Lee ES, Hu G, Kang Y: The miR-200 family inhibits epithelial-
mesenchymal transition and cancer cell migration by direct targeting of
E-cadherin transcriptional repressors ZEB1 and ZEB2. J Biol Chem 2008,
283(22):1491014914.
53. Huber MA, Kraut N, Beug H: Molecular requirements for epithelial-
mesenchymal transition during tumor progression. Curr Opin Cell Biol
2005, 17(5):548558.
54. Park SM, Gaur AB, Lengyel E, Peter ME: The miR-200 family determines the
epithelial phenotype of cancer cells by targeting the E-cadherin
repressors ZEB1 and ZEB2. Genes Dev 2008, 22(7):894907.
55. Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA,
Khew-Goodall Y, Goodall GJ: The miR-200 family and miR-205 regulate
epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell
Biol 2008, 10(5):593601.
56. Gregory PA, Bracken CP, Bert AG, Goodall GJ: MicroRNAs as regulators of
epithelial-mesenchymal transition. Cell Cycle 2008, 7(20):31123118.
57. Kong D, Li Y, Wang Z, Banerjee S, Ahmad A, Kim HR, Sarkar FH: miR-200
regulates PDGF-D-mediated epithelial-mesenchymal transition, adhesion,
and invasion of prostate cancer cells. Stem Cells 2009, 27(8):17121721.
58. Imai T, Horiuchi A, Shiozawa T, Osada R, Kikuchi N, Ohira S, Oka K, Konishi I:
Elevated expression of E-cadherin and alpha-, beta-, and gamma-
catenins in metastatic lesions compared with primary epithelial ovarian
carcinomas. Hum Pathol 2004, 35(12):14691476.
59. Leskela S, Leandro-Garcia LJ, Mendiola M, Barriuso J, Inglada-Perez L, Munoz
I, Martinez-Delgado B, Redondo A, de Santiago J, Robledo M, et al:The
miR-200 family controls beta-tubulin III expression and is associated with
paclitaxel-based treatment response and progression-free survival in
ovarian cancer patients. Endocr Relat Cancer 2011, 18(1):8595.
60. Rosell R, Scagliotti G, Danenberg KD, Lord RV, Bepler G, Novello S, Cooc J,
Crino L, Sanchez JJ, Taron M, et al:Transcripts in pretreatment biopsies
from a three-arm randomized trial in metastatic non-small-cell lung
cancer. Oncogene 2003, 22(23):35483553.
61. Seve P, Mackey J, Isaac S, Tredan O, Souquet PJ, Perol M, Lai R, Voloch A,
Dumontet C: Class III beta-tubulin expression in tumor cells predicts
response and outcome in patients with non-small cell lung cancer
receiving paclitaxel. Mol Cancer Ther 2005, 4(12):20012007.
62. Seve P, Dumontet C: Is class III beta-tubulin a predictive factor in
patients receiving tubulin-binding agents? Lancet Oncol 2008,
9(2):168175.
63. Koh Y, Kim TM, Jeon YK, Kwon TK, Hah JH, Lee SH, Kim DW, Wu HG, Rhee
CS, Sung MW, et al:Class III beta-tubulin, but not ERCC1, is a strong
predictive and prognostic marker in locally advanced head and neck
squamous cell carcinoma. Ann Oncol 2009, 20(8):14141419.
64. Ferrandina G, Zannoni GF, Martinelli E, Paglia A, Gallotta V, Mozzetti S,
Scambia G, Ferlini C: Class III beta-tubulin overexpression is a marker of
poor clinical outcome in advanced ovarian cancer patients. Clin Cancer
Res 2006, 12(9):27742779.
65. Kavallaris M, Kuo DY, Burkhart CA, Regl DL, Norris MD, Haber M, Horwitz SB:
Taxol-resistant epithelial ovarian tumors are associated with altered
expression of specific beta-tubulin isotypes. J Clin Invest 1997,
100(5):12821293.
66. Mozzetti S, Ferlini C, Concolino P, Filippetti F, Raspaglio G, Prislei S, Gallo D,
Martinelli E, Ranelletti FO, Ferrandina G, et al:Class III beta-tubulin
overexpression is a prominent mechanism of paclitaxel resistance in
ovarian cancer patients. Clin Cancer Res 2005, 11(1):298305.
67. Umezu T, Shibata K, Kajiyama H, Terauchi M, Ino K, Nawa A, Kikkawa F:
Taxol resistance among the different histological subtypes of ovarian
cancer may be associated with the expression of class III beta-tubulin.
Int J Gynecol Pathol 2008, 27(2):207212.
68. Roush S, Slack FJ: The let-7 family of microRNAs. Trends Cell Biol 2008,
18(10):505516.
69. Pasquinelli AE, Reinhart BJ, Slack F, Martindale MQ, Kuroda MI, Maller B,
Hayward DC, Ball EE, Degnan B, Muller P, et al:Conservation of the
sequence and temporal expression of let-7 heterochronic regulatory
RNA. Nature 2000, 408(6808):8689.
70. Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Endoh H,
Harano T, Yatabe Y, Nagino M, Nimura Y, et al:Reduced expression of the
let-7 microRNAs in human lung cancers in association with shortened
postoperative survival. Cancer Res 2004, 64(11):37533756.
71. Yang N, Kaur S, Volinia S, Greshock J, Lassus H, Hasegawa K, Liang S,
Leminen A, Deng S, Smith L, et al:MicroRNA microarray identifies Let-7i
as a novel biomarker and therapeutic target in human epithelial ovarian
cancer. Cancer Res 2008, 68(24):1030710314.
72. Johnson SM, Grosshans H, Shingara J, Byrom M, Jarvis R, Cheng A, Labourier
E, Reinert KL, Brown D, Slack FJ: RAS is regulated by the let-7 microRNA
family. Cell 2005, 120(5):635647.
73. Bussing I, Slack FJ, Grosshans H: let-7 microRNAs in development, stem
cells and cancer. Trends Mol Med 2008, 14(9):400409.
74. Johnson CD, Esquela-Kerscher A, Stefani G, Byrom M, Kelnar K, Ovcharenko
D, Wilson M, Wang X, Shelton J, Shingara J, et al:The let-7 microRNA
represses cell proliferation pathways in human cells. Cancer Res 2007,
67(16):77137722.
75. Schultz J, Lorenz P, Gross G, Ibrahim S, Kunz M: MicroRNA let-7b targets
important cell cycle molecules in malignant melanoma cells and
interferes with anchorage-independent growth. Cell Res 2008,
18(5):549557.
76. Wang Y, Hu X, Greshock J, Shen L, Yang X, Shao Z, Liang S, Tanyi JL, Sood
AK, Zhang L: Genomic DNA Copy-Number Alterations of the let-7 Family
in Human Cancers. PLoS One 2012, 7(9):e44399.
77. Chen GQ, Zhao ZW, Zhou HY, Liu YJ, Yang HJ: Systematic analysis of
microRNA involved in resistance of the MCF-7 human breast cancer cell
to doxorubicin. Med Oncol 2010, 27(2):406415.
78. Blower PE, Chung JH, Verducci JS, Lin S, Park JK, Dai Z, Liu CG, Schmittgen
TD, Reinhold WC, Croce CM, et al:MicroRNAs modulate the
chemosensitivity of tumor cells. Mol Cancer Ther 2008, 7(1):19.
79. Weidhaas JB, Babar I, Nallur SM, Trang P, Roush S, Boehm M, Gillespie E,
Slack FJ: MicroRNAs as potential agents to alter resistance to cytotoxic
anticancer therapy. Cancer Res 2007, 67(23):1111111116.
80. Boyerinas B, Park SM, Murmann AE, Gwin K, Montag AG, Zillhardt M, Hua YJ,
Lengyel E, Peter ME: Let-7 modulates acquired resistance of ovarian
cancer to Taxanes via IMP-1-mediated stabilization of multidrug
resistance 1. Int J Cancer 2012, 130(8):17871797.
81. Lu L, Schwartz P, Scarampi L, Rutherford T, Canuto EM, Yu H, Katsaros D:
MicroRNA let-7a: a potential marker for selection of paclitaxel in ovarian
cancer management. Gynecol Oncol 2011, 122(2):366371.
82. Corney DC, Hwang CI, Matoso A, Vogt M, Flesken-Nikitin A, Godwin AK,
Kamat AA, Sood AK, Ellenson LH, Hermeking H, et al:Frequent
downregulation of miR-34 family in human ovarian cancers. Clin Cancer
Res 2010, 16(4):11191128.
83. Nagaraja AK, Creighton CJ, Yu Z, Zhu H, Gunaratne PH, Reid JG, Olokpa E,
Itamochi H, Ueno NT, Hawkins SM, et al:A link between mir-100 and
FRAP1/mTOR in clear cell ovarian cancer. Mol Endocrinol 2010,
24(2):447463.
84. Peng DX, Luo M, Qiu LW, He YL, Wang XF: Prognostic implications of
microRNA-100 and its functional roles in human epithelial ovarian
cancer. Oncol Rep 2012, 27(4):12381244.
85. Mabuchi S, Kawase C, Altomare DA, Morishige K, Sawada K, Hayashi M,
Tsujimoto M, Yamoto M, Klein-Szanto AJ, Schilder RJ, et al:mTOR is a
promising therapeutic target both in cisplatin-sensitive and cisplatin-
resistant clear cell carcinoma of the ovary. Clin Cancer Res 2009,
15(17):54045413.
86. Creighton CJ, Fountain MD, Yu Z, Nagaraja AK, Zhu H, Khan M, Olokpa E,
Zariff A, Gunaratne PH, Matzuk MM, et al:Molecular profiling uncovers a
p53-associated role for microRNA-31 in inhibiting the proliferation of
serous ovarian carcinomas and other cancers. Cancer Res 2010,
70(5):19061915.
87. Ueda T, Volinia S, Okumura H, Shimizu M, Taccioli C, Rossi S, Alder H, Liu CG,
Oue N, Yasui W, et al:Relation between microRNA expression and
progression and prognosis of gastric cancer: a microRNA expression
analysis. Lancet Oncol 2010, 11(2):136146.
88. Penna E, Orso F, Cimino D, Tenaglia E, Lembo A, Quaglino E, Poliseno L,
Haimovic A, Osella-Abate S, De Pitta C, et al:microRNA-214 contributes to
melanoma tumour progression through suppression of TFAP2C.
EMBO J 2011, 30(10):19902007.
89. Volinia S, Calin GA, Liu CG, Ambs S, Cimmino A, Petrocca F, Visone R, Iorio
M, Roldo C, Ferracin M, et al:A microRNA expression signature of human
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 10 of 11
http://www.ovarianresearch.com/content/5/1/44
solid tumors defines cancer gene targets. Proc Natl Acad Sci U S A 2006,
103(7):22572261.
90. Blenkiron C, Goldstein LD, Thorne NP, Spiteri I, Chin SF, Dunning MJ,
Barbosa-Morais NL, Teschendorff AE, Green AR, Ellis IO, et al:MicroRNA
expression profiling of human breast cancer identifies new markers of
tumor subtype. Genome Biol 2007, 8(10):R214.
91. Qiang R, Wang F, Shi LY, Liu M, Chen S, Wan HY, Li YX, Li X, Gao SY, Sun BC,
et al:Plexin-B1 is a target of miR-214 in cervical cancer and promotes
the growth and invasion of HeLa cells. Int J Biochem Cell Biol 2011,
43(4):632641.
92. Testa JR, Bellacosa A: AKT plays a central role in tumorigenesis. Proc Natl
Acad Sci U S A 2001, 98(20):1098310985.
93. Yin G, Chen R, Alvero AB, Fu HH, Holmberg J, Glackin C, Rutherford T, Mor
G: TWISTing stemness, inflammation and proliferation of epithelial
ovarian cancer cells through MIR199A2/214. Oncogene 2010,
29(24):35453553.
94. Lee MS, Lowe GN, Strong DD, Wergedal JE, Glackin CA: TWIST, a basic
helix-loop-helix transcription factor, can regulate the human osteogenic
lineage. J Cell Biochem 1999, 75(4):566577.
95. Lee MS, Lowe G, Flanagan S, Kuchler K, Glackin CA: Human Dermo-1 has
attributes similar to twist in early bone development. Bone 2000,
27(5):591602.
96. Bialek P, Kern B, Yang X, Schrock M, Sosic D, Hong N, Wu H, Yu K, Ornitz
DM, Olson EN, et al:A twist code determines the onset of osteoblast
differentiation. Dev Cell 2004, 6(3):423435.
97. Ota MS, Loebel DA, O'Rourke MP, Wong N, Tsoi B, Tam PP: Twist is
required for patterning the cranial nerves and maintaining the viability
of mesodermal cells. Dev Dyn 2004, 230(2):216228.
98. Li Z, Lu J, Sun M, Mi S, Zhang H, Luo RT, Chen P, Wang Y, Yan M, Qian Z,
et al:Distinct microRNA expression profiles in acute myeloid leukemia
with common translocations. Proc Natl Acad Sci U S A 2008,
105(40):1553515540.
99. Xu G, Zhong Y, Munir S, Yang BB, Tsang BK, Peng C: Nodal induces
apoptosis and inhibits proliferation in human epithelial ovarian cancer
cells via activin receptor-like kinase 7. J Clin Endocrinol Metab 2004,
89(11):55235534.
100. Li Y, Tan W, Neo TW, Aung MO, Wasser S, Lim SG, Tan TM: Role of the miR-
106b-25 microRNA cluster in hepatocellular carcinoma. Cancer Sci 2009,
100(7):12341242.
101. Fang L, Deng Z, Shatseva T, Yang J, Peng C, Du WW, Yee AJ, Ang LC, He C,
Shan SW, et al:MicroRNA miR-93 promotes tumor growth and
angiogenesis by targeting integrin-beta8. Oncogene 2011, 30(7):806821.
102. Fu X, Tian J, Zhang L, Chen Y, Hao Q: Involvement of microRNA-93, a new
regulator of PTEN/Akt signaling pathway, in regulation of
chemotherapeutic drug cisplatin chemosensitivity in ovarian cancer
cells. FEBS Lett 2012, 586(9):12791286.
103. Altomare DA, Wang HQ, Skele KL, De Rienzo A, Klein-Szanto AJ, Godwin AK,
Testa JR: AKT and mTOR phosphorylation is frequently detected in
ovarian cancer and can be targeted to disrupt ovarian tumor cell
growth. Oncogene 2004, 23(34):58535857.
104. Si ML, Zhu S, Wu H, Lu Z, Wu F, Mo YY: miR-21-mediated tumor growth.
Oncogene 2007, 26(19):27992803.
105. Slaby O, Svoboda M, Fabian P, Smerdova T, Knoflickova D, Bednarikova M,
Nenutil R, Vyzula R: Altered expression of miR-21, miR-31, miR-143 and
miR-145 is related to clinicopathologic features of colorectal cancer.
Oncology 2007, 72(56):397402.
106. Lou Y, Yang X, Wang F, Cui Z, Huang Y: MicroRNA-21 promotes the cell
proliferation, invasion and migration abilities in ovarian epithelial
carcinomas through inhibiting the expression of PTEN protein. Int J Mol
Med 2010, 26(6):819827.
107. Polytarchou C, Iliopoulos D, Hatziapostolou M, Kottakis F, Maroulakou I,
Struhl K, Tsichlis PN: Akt2 regulates all Akt isoforms and promotes
resistance to hypoxia through induction of miR-21 upon oxygen
deprivation. Cancer Res 2011, 71(13):47204731.
108. Li J, Liang S, Yu H, Zhang J, Ma D, Lu X: An inhibitory effect of miR-22 on
cell migration and invasion in ovarian cancer. Gynecol Oncol 2010,
119(3):543548.
109. Kim TH, Kim YK, Kwon Y, Heo JH, Kang H, Kim G, An HJ: Deregulation of
miR-519a, 153, and 485-5p and its clinicopathological relevance in
ovarian epithelial tumours. Histopathology 2010, 57(5):734743.
110. Lu L, Katsaros D, de la Longrais IA, Sochirca O, Yu H: Hypermethylation of
let-7a-3 in epithelial ovarian cancer is associated with low insulin-like
growth factor-II expression and favorable prognosis. Cancer Res 2007,
67(21):1011710122.
111. Hu X, Macdonald DM, Huettner PC, Feng Z, El Naqa IM, Schwarz JK,
Mutch DG, Grigsby PW, Powell SN, Wang X: A miR-200 microRNA cluster
as prognostic marker in advanced ovarian cancer. Gynecol Oncol 2009,
114(3):457464.
112. Taylor DD, Gercel-Taylor C: MicroRNA signatures of tumor-derived
exosomes as diagnostic biomarkers of ovarian cancer. Gynecol Oncol
2008, 110(1):1321.
113. Shell S, Park SM, Radjabi AR, Schickel R, Kistner EO, Jewell DA, Feig C,
Lengyel E, Peter ME: Let-7 expression defines two differentiation stages
of cancer. Proc Natl Acad Sci U S A 2007, 104(27):1140011405.
114. Garzon R, Marcucci G, Croce CM: Targeting microRNAs in cancer:
rationale, strategies and challenges. Nat Rev Drug Discov 2010,
9(10):775789.
115. Lu Y, Xiao J, Lin H, Bai Y, Luo X, Wang Z, Yang B: A single anti-microRNA
antisense oligodeoxyribonucleotide (AMO) targeting multiple microRNAs
offers an improved approach for microRNA interference. Nucleic Acids Res
2009, 37(3):e24.
116. Trang P, Medina PP, Wiggins JF, Ruffino L, Kelnar K, Omotola M, Homer R,
Brown D, Bader AG, Weidhaas JB, et al:Regression of murine lung tumors
by the let-7 microRNA. Oncogene 2010, 29(11):15801587.
117. Trang P, Wiggins JF, Daige CL, Cho C, Omotola M, Brown D, Weidhaas JB,
Bader AG, Slack FJ: Systemic delivery of tumor suppressor microRNA
mimics using a neutral lipid emulsion inhibits lung tumors in mice.
Mol Ther 2011, 19(6):11161122.
118. Pramanik D, Campbell NR, Karikari C, Chivukula R, Kent OA, Mendell JT,
Maitra A: Restitution of tumor suppressor microRNAs using a systemic
nanovector inhibits pancreatic cancer growth in mice. Mol Cancer Ther
2011, 10(8):14701480.
119. Suzuki HI, Yamagata K, Sugimoto K, Iwamoto T, Kato S, Miyazono K:
Modulation of microRNA processing by p53. Nature 2009,
460(7254):529533.
120. Liu C, Kelnar K, Liu B, Chen X, Calhoun-Davis T, Li H, Patrawala L, Yan H,
Jeter C, Honorio S, et al:The microRNA miR-34a inhibits prostate cancer
stem cells and metastasis by directly repressing CD44. Nat Med 2011,
17(2):211215.
121. Melo S, Villanueva A, Moutinho C, Davalos V, Spizzo R, Ivan C, Rossi S,
Setien F, Casanovas O, Simo-Riudalbas L, et al:Small molecule enoxacin is
a cancer-specific growth inhibitor that acts by enhancing TAR RNA-
binding protein 2-mediated microRNA processing. Proc Natl Acad Sci
USA2011, 108(11):43944399.
122. Shan G, Li Y, Zhang J, Li W, Szulwach KE, Duan R, Faghihi MA, Khalil AM,
Lu L, Paroo Z, et al:A small molecule enhances RNA interference and
promotes microRNA processing. Nat Biotechnol 2008, 26(8):933940.
123. Jeon HM, Sohn YW, Oh SY, Kim SH, Beck S, Kim S, Kim H: ID4 imparts
chemoresistance and cancer stemness to glioma cells by derepressing
miR-9*-mediated suppression of SOX2. Cancer Res 2011, 71(9):34103421.
124. Cittelly DM, Das PM, Salvo VA, Fonseca JP, Burow ME, Jones FE: Oncogenic
HER2{Delta}16 suppresses miR-15a/16 and deregulates BCL-2 to
promote endocrine resistance of breast tumors. Carcinogenesis 2010,
31(12):20492057.
125. Hwang JH, Voortman J, Giovannetti E, Steinberg SM, Leon LG, Kim YT, Funel
N, Park JK, Kim MA, Kang GH, et al:Identification of microRNA-21 as a
biomarker for chemoresistance and clinical outcome following adjuvant
therapy in resectable pancreatic cancer. PLoS One 2010, 5(5):e10630.
doi:10.1186/1757-2215-5-44
Cite this article as: Zaman et al.:Current status and implications of
microRNAs in ovarian cancer diagnosis and therapy. Journal of Ovarian
Research 2012 5:44.
Zaman et al. Journal of Ovarian Research 2012, 5:44 Page 11 of 11
http://www.ovarianresearch.com/content/5/1/44
... The most common women's reproductive diseases include polycystic ovary syndrome (PCOS) [15], endometriosis (EMs) [16,17], uterine leiomyomas (ULs) [18], and ovarian cancer (OC) [12,19]. Since these diseases do not exhibit obvious symptoms at the onset, they are typically diagnosed during the advanced stage [20]. The mechanistic diagram of ovarian/uterine dysfunctions was shown in Fig. 1. ...
... Besides these novel therapeutic methods, multidisciplinary approaches with different specialists are needed to treat OC to determine the treatment protocol for each patient based on the patient's characteristics and the tumor state [132,133]. On the other hand, the symptoms of OC are often absent or vague at the onset of the disease, and due to the lack of early diagnostic methods and reliable molecular markers, most patients are diagnosed at an advanced stage, and only 20% of them are diagnosed at stage I of the disease [20]. ...
Article
Full-text available
Background One of the main health issues that can affect women's health is reproductive diseases, such as polycystic ovary syndrome (PCOS), endometriosis (EMs), uterine leiomyomas (ULs), and ovarian cancer (OC). Although these diseases are very common, we do not have a complete understanding of their underlying cellular and molecular mechanisms. It is important to mention that the majority of patients are diagnosed with these diseases at later stages because of the absence of early diagnostic techniques and dependable molecular indicators. Hence, it is crucial to discover novel and non-invasive biomarkers that have prognostic, diagnostic and therapeutic capabilities. MiRNAs, also known as microRNAs, are small non-coding RNAs that play a crucial role in regulating gene expression at the post-transcriptional level. They are short in length, typically consisting of around 22 nucleotides, and are highly conserved across species. Numerous studies have shown that miRNAs are expressed differently in various diseases and can act as either oncogenes or tumor suppressors. Methods The author conducted a comprehensive review of all the pertinent papers available in web of science, PubMed, Google Scholar, and Scopus databases. Results We achieved three goals: providing readers with better information, enhancing search results, and making peer review easier. Conclusions This review focuses on the investigation of miRNAs and their involvement in various reproductive disorders in women, including their molecular targets. Additionally, it explores the role of miRNAs in the development and progression of these disorders.
... MiRNA-100, a tumor suppressor that inhibits mTOR, is also down-regulated in OC and is associated with shorter overall survival in patients with advanced-stage OC [22,36]. In addition, miR-100 inhibits the expression of the proto-oncogene PLK1 (Polo-like kinase-1) in OC [37]. MiR-31, as a transcriptional repressor mediated by p53 and as a tumor suppressor in OC, inhibits the expression of cell cycle regulators such as E2F2 and STK40 [38]. ...
Article
Full-text available
Ovarian cancer, a perilous form of cancer affecting the female reproductive system, exhibits intricate communication networks that contribute to its progression. This study aims to identify crucial molecular abnormalities linked to the disease to enhance diagnostic and therapeutic strategies. In particular, we investigate the role of microRNAs (miRNAs) as diagnostic biomarkers and explore their potential in treating ovarian cancer. By targeting miRNAs, which can influence multiple pathways and genes, substantial therapeutic benefits can be attained. In this review we want to shed light on the promising application of miRNA-based interventions and provide insights into the specific miRNAs implicated in ovarian cancer pathogenesis.
... MiR-200 and let-7 -two families of miRNAs -are often dysregulated in EOC and have been associated with a dismal outcome. They have been linked to the epithelial-to-mesenchymal transition, a cellular change correlated with tumour aggressiveness, invasion, and resistance to chemotherapy [98]. According to Zhang et al., a high proportion of genomic loci containing miRNA genes had DNA copy number alterations in ovarian cancer (37.1 %), breast cancer (72.8%), and melanoma (85.9%) that can induce gene expression changes. ...
Article
Ovarian cancer has become the largest cause of gynaecological cancer-related mortality. It is typically diagnosed at a late stage and has no effective screening strategy. Ovarian cancer is a highly heterogeneous disease that can be subdivided into several molecular subsets. As a result of a greater understanding of molecular pathways involved in carcinogenesis and tumor growth, targeted agents have been approved or are in several stages of development. Poly(ADP-ribose) polymerase (PARP) inhibitors and the anti-vascular endothelial growth factor (VEGF)-A antibodies are two types of approved and most effective targeted drugs for ovarian cancer at present. With the success of bevacizumab, tyrosine kinase inhibitors which could target alternate angiogenic pathways are being studied. Furthermore, many treatments targeting the PI3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathways, are being developed or are already in clinical studies. MicroRNAs have also become novel biomarkers for the therapy and clinical diagnosis of ovarian cancer. This manuscript reviews the molecular, preclinical and clinical evidence supporting the targeting of growth-dependent pathways in ovarian cancer and assesses current data related to targeted treatments beyond PARP inhibitors.
... Disorders of miR-21 regulation occur in a variety of cancers, such as breast cancer, colon cancer, lung cancer, prostate cancer, pancreatic cancer, gastric cancer, ovarian cancer, esophageal cancer, and liver cancer, which were used as a qualitative and quantitative marker for potential cancer detection, as well as potential targets for treatment [37]. Certain miRNAs, such as miR-200 family, let-7 family, miR-21, miR-214 and miR-100, have the potential diagnostic and prognostic value in ovarian cancer through the special signaling pathway [38]. MiR-21 regulates the proliferation and apoptosis of ovarian cancer cells through the PTEN/PI3K/AKT pathway [39] and the jagged-1 pathway [40], regulating cell invasion, migration and colony formation by the miR-21/Wnt/ CD44v6 pathway [41]. ...
Article
Full-text available
Objective There have been a variety of published studies on the expression of serum miR-21 in patients with ovarian cancer associated with the diagnostic value of ovarian cancer, but the conclusions are not clearly elucidated. This study aims to evaluate the value of serum miR-21 expression in the diagnosis of patients with ovarian cancer by meta-analysis. Methods Databases, such as PubMed, Embase, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI), and China WanFang, were searched for relevant studies upon the correlation between the expression of serum miR-21 and the diagnostic value of ovarian cancer from inception to March 7, 2022. Statistical analysis was performed using Stata 15.0 software. The pooled sensitivity, specificity, diagnostic odds ratio (DOR), positive likelihood ratio (PLR), and negative likelihood ratio (NLR) were calculated. The meta-regression analysis and subgroup analysis were used to explore the sources of heterogeneity. The Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) system was used to evaluate the quality of the included literature. Results A total of 6 articles were included in the meta-analysis. The results showed that the pooled sensitivity, specificity, PLR, NLR, and DOR were 0.81 (95%CI: 0.73–0.88), 0.82 (95%CI: 0.75–0.87), 4.51 (95%CI: 3.22–6.31), 0.23 (95%CI: 0.16–0.33), and 19.87 (95% CI: 11.27–35.03), respectively. The area under the summary receiver operating characteristic curve was 0.89 (95%CI: 0.85–0.91). No significant publication bias was found ( P > 0.05). Conclusion Serum miR-21 has a good diagnostic value for ovarian cancer, which can be an ideal diagnostic biomarker for ovarian cancer. However, we should gingerly use miR-21 as a diagnostic reference standard, due to the limited number of included studies and heterogeneity.
... For this, miR-153 binds to the 3'UTR region of SET7/ZEB2, and promotes its mRNA degradation. By downregulating SET7 and ZEB2, miR-153 inhibits the proliferation in EOC (Epithelial ovarian cancer) cells and suppresses EMT (Zaman et al., 2012). miR-153 also targets the AKT1 in EOC to inhibit proliferation, migration, and colony formation since PI3K/AKT pathway inhibition and EOC tumor growth is negatively correlated (Li et al., 2017). ...
Article
Full-text available
MicroRNA-153 (miR-153), belongs to a class of small non-coding RNA. It is a critical regulator of gene expression at the post-transcriptional level which interacts with the functional mRNA at 3’UTR region and suppresses the expression of the mRNA. More recently, it has become apparent that changes in the miR-153 expression lead to invasion, metastasis, angiogenesis and various types of tumor progression. This review summarizes the connection between dysregulation of miR-153 and various types of cancer progression. miR-153 regulates various signaling pathways to inhibit the proliferation and induce apoptosis in the cancer cell and also show synergistic activity with anticancer drugs. In addition to this, the oncogenic behavior of miR-153 and their use as a potential biomarker in cancer was also reviewed.
Article
Full-text available
Ovarian cancer (OC) is a fatal gynecological disease that is often diagnosed at later stages due to its asymptomatic nature and the absence of efficient early-stage biomarkers. Previous studies have identified genes with abnormal expression in OC that couldn’t be explained by methylation or mutation, indicating alternative mechanisms of gene regulation. Recent advances in human transcriptome studies have led to research on non-coding RNAs (ncRNAs) as regulators of cancer gene expression. Long non-coding RNAs (lncRNAs), a class of ncRNAs with a length greater than 200 nucleotides, have been identified as crucial regulators of physiological processes and human diseases, including cancer. Dysregulated lncRNA expression has also been found to play a crucial role in ovarian carcinogenesis, indicating their potential as novel and non-invasive biomarkers for improving OC management. However, despite the discovery of several thousand lncRNAs, only one has been approved for clinical use as a biomarker in cancer, highlighting the importance of further research in this field. In addition to their potential as biomarkers, lncRNAs have been implicated in modulating chemoresistance, a major problem in OC. Several studies have identified altered lncRNA expression upon drug treatment, further emphasizing their potential to modulate chemoresistance. In this review, we highlight the characteristics of lncRNAs, their function, and their potential to serve as tumor markers in OC. We also discuss a few databases providing detailed information on lncRNAs in various cancer types. Despite the promising potential of lncRNAs, further research is necessary to fully understand their role in cancer and develop effective strategies to combat this devastating disease.
Article
Full-text available
Non-coding RNAs with a length of 22-24 nt are known as microRNAs (miRNAs or miRs), which are critical regulators of protein translation. Over the past 10 years, the roles of miRNAs have been extensively investigated in several human cancer types. There is evidence to indicate that miRNAs regulate gene expression by concentrating on a number of substances that have an impact on the physiology and development of cancer cells. Thus, miRNAs as regarded as effective targets for further studies on the design of novel therapeutic strategies. Hepatocellular carcinoma, breast, prostate, and ovarian cancer are only a few of the cancers that miR-124 suppresses. Furthermore, it has been shown that miR-124 is linked to the development and aggressive spread of malignancies. The aim of the present review was to clarify and highlight the role of miR-124 in the development and progression of cancer, emphasizing recent research illustrating how miR-124 has been used as a therapeutic agent against cancer, as well as the diagnostic potential, regulatory mechanisms and clinical application of miR-124.
Chapter
Chapter 3 through Chap. 11 introduces the current status of clinical applications of polypharmacology to disease therapy. This chapter will describe the applications of polypharmacology to drug discovery, mainly from the theoretical account. Specifically, it will explain the theoretical basis for the rational design of new drugs of the “single agent on multiple targets for single disease” category, or the rational design of MTD drugs. Seven essential strategies for MTD drug discovery, including sequence-based epigenetic drugs, sequence-based small-molecule drugs, function-based epigenetic drugs, function-based small-molecule drugs, function-based macromolecule drugs, naturally occurring MTD drugs, and repurposed MTD drugs, will be described in detail with real examples. The contents of this chapter lay the foundation for better understanding of Section II chapters on the methodologies of MTD polypharmacology-based drug discovery and development. Several new concepts on the rational design and discovery of MTDs are proposed by us and presented in this chapter.
Article
Full-text available
MicroRNAs (miRNAs) are short, noncoding, single-stranded RNA molecules that regulate gene expression at the post-transcriptional level by binding to mRNAs. miRNAs affect the course of processes of fundamental importance for the proper functioning of the organism. These processes include cell division, proliferation, differentiation, cell apoptosis and the formation of blood vessels. Altered expression of individual miRNAs has been shown in numerous cancers, which may indicate the oncogenic or suppressor potential of the molecules in question. This paper discusses the current knowledge about the possibility of using miRNA as a diagnostic marker and a potential target in modern anticancer therapies.
Article
Full-text available
A large number of tiny noncoding RNAs have been cloned and named microRNAs (miRs). Recently, we have reported that miR-15a and miR-16a, located at 13q14, are frequently deleted andor down-regulated in patients with B cell chronic lymphocytic leuke-mia, a disorder characterized by increased survival. To further investigate the possible involvement of miRs in human cancers on a genome-wide basis, we have mapped 186 miRs and compared their location to the location of previous reported nonrandom genetic alterations. Here, we show that miR genes are frequently located at fragile sites, as well as in minimal regions of loss of heterozygosity, minimal regions of amplification (minimal ampli-cons), or common breakpoint regions. Overall, 98 of 186 (52.5%) of miR genes are in cancer-associated genomic regions or in fragile sites. Moreover, by Northern blotting, we have shown that several miRs located in deleted regions have low levels of expression in cancer samples. These data provide a catalog of miR genes that may have roles in cancer and argue that the full complement of miRs in a genome may be extensively involved in cancers.
Article
Full-text available
Question: What is the optimal chemotherapy treatment for women with recurrent ovarian cancer who have previously received platinum-based chemotherapy? Perspectives: Currently, standard primary therapy for advanced disease involves a combination of maximal cytoreductive surgery and chemotherapy with carboplatin plus paclitaxel or with carboplatin alone. Despite initial high response rates, a large proportion of patients relapse, resulting in a therapeutic challenge. Because these patients are not curable, the goal of therapy becomes improvement in both quality and length of life. The search has therefore been to find active agents for women with recurrent disease following platinum-based chemotherapy. Outcomes: Outcomes of interest included any combination of tumour response rate, progression-free survival, overall survival, adverse events, and quality of life. Methodology: The medline, embase, and Cochrane Library databases were systematically searched for primary articles and practice guidelines. The resulting evidence informed the development of clinical practice recommendations. The systematic review and recommendations were approved by the Report Approval Panel of the Program in Evidence-Based Care, and by the Gynecology Cancer Disease Site Group (dsg). The practice guideline was externally reviewed by a sample of practitioners from Ontario, Canada. Results: Thirteen randomized trials compared various chemotherapy regimens for patients with recurrent ovarian cancer. In five of the thirteen trials in which 100% of patients were considered sensitive to platinum-containing chemotherapy, further platinum-based combination chemotherapy significantly improved response rates (two trials), progression-free survival (four trials), and overall survival (three trials) when compared with single-agent chemotherapy involving carboplatin or paclitaxel. Only two of these randomized trials compared the same chemotherapy regimens: carboplatin alone versus the combination of carboplatin and paclitaxel. Both trials were consistent in reporting improved survival outcomes with the combination of carboplatin and paclitaxel. In one trial, the combination of carboplatin and gemcitabine resulted in significantly higher response rates and improved progression-free survival when compared with carboplatin alone. Median survival with carboplatin alone ranged from 17 months to 24 months in four trials. In eight of the thirteen trials in which 35%–100% of patients had platinum-refractory or -resistant disease, one trial reported a statistically significant 2-month improvement in overall survival with liposomal doxorubicin as compared with topotecan (15 months vs. 13 months, p = 0.038; hazard ratio: 1.23; 95% confidence interval: 1.01 to 1.50). In that trial, because of the limited clinical benefit and the unusual finding that a survival difference emerged only after a year of treatment with no corresponding improvement in the rate of response or of progression-free survival, the authors concluded that further confirmation by results from randomized trials were needed to establish the superiority of one agent over another in their trial. In one trial, topotecan was superior to treosulphan in patient progression-free survival by a span of approximately 2 months (5.4 months vs. 3.0 months, p < 0.001). Toxicity was reported in all of the randomized trials, and although data on adverse events varied by treatment regimen, the observed adverse events correlated with known toxicity profiles. As expected, combination chemotherapy was associated with higher rates of adverse events. Practice Guideline: Target Population This clinical recommendation applies to women with recurrent epithelial ovarian cancer who have previously received platinum-based chemotherapy. Of specific interest are women who have previously shown sensitivity to platinum therapy and those who previously were refractory or resistant to platinum-based chemotherapy. As a general categorization within what is actually a continuum, “platinum sensitivity” refers to disease recurrence 6 months or more after prior platinum-containing chemotherapy, and “platinum resistance” refers to a response to platinum-based chemotherapy followed by relapse less than 6 months after chemotherapy is stopped. “Platinum-refractory disease” refers to a lack of response or to progression while on platinum-based chemotherapy. Recommendations Although the body of evidence that informs the clinical recommendations is based on randomized trial data, those data are incomplete. Based on the available data and expert consensus opinion, the Gynecology Cancer dsg makes these recommendations: Systemic therapy for recurrent ovarian cancer is not curative. It is therefore recognized that each patient must be individually assessed to determine optimal therapy in terms of recurrence, sensitivity to platinum, toxicity, ease of administration, and patient preference. All suitable patients should be offered the opportunity to participate in randomized trials, if available. In the absence of contraindications, combination platinum-based chemotherapy should be considered for patients with prior sensitivity to platinum-containing chemotherapy. As compared with carboplatin alone, the combination of carboplatin and paclitaxel significantly improved both progression-free and overall survival. If combination platinum-based chemotherapy is not indicated, then a single platinum agent should be considered. Carboplatin has demonstrated efficacy across trials and has a manageable toxicity profile. If a single platinum agent is not being considered, then monotherapy with paclitaxel, topotecan, or pegylated liposomal doxorubicin are seen as reasonable treatment options. Some patients may be repeatedly sensitive to treatment and may benefit from multiple lines of chemotherapy. For patients with platinum-refractory or platinum-resistant disease, the goals of treatment should be to improve quality of life by extending the symptom-free interval, by reducing symptom intensity, and by increasing progression-free interval, and, if possible, to prolong life. With non-platinum agents, monotherapy should be considered because no advantage appears to accrue to the use of non-platinum-containing combination chemotherapy in this group of patients. Single-agent paclitaxel, topotecan, or pegylated liposomal doxorubicin have demonstrated activity in this patient population and are reasonable treatment options. No evidence either supports or refutes the use of more than one line of chemotherapy in patients with platinum-refractory or platinum-resistant recurrence. Many treatment options have shown modest response rates, but their benefits over best supportive care have not been studied in clinical trials.
Article
Full-text available
Background: Despite considerable improvement in the treatment of advanced ovarian cancer, the optimization of efficacy and tolerability remains an important issue. Therefore, we performed a randomized, phase III non-inferiority trial comparing paclitaxel plus cisplatin (PT) with paclitaxel plus carboplatin (TC) in patients with advanced ovarian cancer. Methods: A total of 798 patients with International Federation of Gynecology and Obstetrics stage IIB–IV were randomly assigned to receive six courses of either PT or TC at 3-week intervals. The primary endpoint was the proportion of patients without progression at 2 years. Secondary endpoints included toxicity, response to treatment, quality of life, and overall and progression-free survival time. Quality of life was evaluated using the European Organization for Research and Treatment of Cancer quality-of-life questionnaire (QLQ)-C30, version 2.0. Survival curves were calculated using the Kaplan–Meier method, and hazard ratios were estimated using the Cox proportional hazards model. Results: The proportion of patients without progression at 2 years was not statistically significantly different between the two treatment arms (40.0% for PT versus 37.5% for TC, difference = 2.5%, one-sided 95% confidence interval [CI] = –∞ to 8.2%). Median progression-free survival time in the TC arm (17.2 months, 95% CI = 15.2 to 19.3 months) and the PT arm (19.1 months, 95% CI = 16.7 to 21.5 months) were also not statistically significantly different; the same was true of median overall survival time (43.3 months, 95% CI = 37.2 to 47.8 months versus 44.1 months, 95% CI = 40.2 to 49.4 months, for the TC and PT arms, respectively). The TC regimen was associated with a higher frequency of hematologic toxicity, but a lower frequency of gastrointestinal and neurologic toxicity, than the PT regimen. Mean global quality-of-life scores at the end of treatment were statistically significantly better in the TC arm than in the PT arm (65.25 versus 51.97, respectively; difference = –13.28, 95% CI = –18.88 to –7.68). Conclusion: The TC regimen achieved comparable efficacy to the PT regimen but was associated with better tolerability and quality of life, and should, therefore, be considered as an important alternative for standard first-line chemotherapy in patients with advanced ovarian cancer.
Article
Epithelial−mesenchymal transition (EMT) is a complex, highly regulated process that is reportedly controlled by hundreds of transcription factors and hundreds of microRNAs. Here we discuss whether the complexity of the process necessitates so many regulators, or whether many ‘false positives’ are reported in the literature. Bracken and Goodall discuss why epithelial–mesenchymal transition (EMT) has so many regulators, but also consider whether many of these may be ‘false positives’.
Article
Basic helix-loop-helix (bHLH) transcription factors have been shown to play an important role in controlling cell type determination and differentiation. TWIST, a member of the bHLH transcription factor family, is involved in the development of mesodermally derived tissue, including the skeleton. We examined the role of human TWIST in osteoblast metabolism using stable expression of sense and antisense TWIST in human osteoblast HSaOS-2 cells. Changes in morphology and osteogenic phenotype characterized these stable clones. Cells that overexpressed TWIST exhibited a spindle shaped morphology, reduced levels of alkaline phosphatase, a reduced proliferation rate, and failed to respond to basic fibroblast growth factor (bFGF). In contrast, those that underexpressed TWIST demonstrated a cuboidal epithelial-like morphology characteristic of differentiated osteoblasts. TWIST antisense cells exhibited increased levels of alkaline phosphatase and type I collagen mRNA, initiated osteopontin mRNA expression, and had a reduced proliferation rate. These results indicate that TWIST overexpressing cells may de-differentiate and remain in an osteoprogenitor-like state, and antisense TWIST cells progress to a more differentiated mature osteoblast-like state. Therefore, the level of TWIST can influence osteogenic gene expression and may act as a master switch in initiating bone cell differentiation by regulating the osteogenic cell lineage. J. Cell. Biochem. 75:566–577, 1999. © 1999 Wiley-Liss, Inc.
Article
These NCCN Guidelines Insights focus on the major updates to the 2013 NCCN Guidelines for Ovarian Cancer. Four updates were selected based on recent important updates in the guidelines and on debate among panel members about recent clinical trials. The topics include 1) intraperitoneal chemotherapy, 2) CA-125 monitoring for ovarian cancer recurrence, 3) surveillance recommendations for less common ovarian histopathologies, and 4) recent changes in therapy for recurrent epithelial ovarian cancer. These NCCN Guidelines Insights also discuss why some recommendations were not made.
Article
Ovarian cancer, especially epithelial ovarian cancer (EOC), which accounts for 90% of ovarian cancer, continues to be the leading cause of death among gynecological malignancies. However, the factors associated with its malignant biological behavior are still poorly understood. Accumulating evidence suggests that microRNAs (miRNAs), regulating diverse biological processes, may play an important role in tumorigenesis and development. miR-21 has been frequently observed to be aberrantly overexpressed in various tumors. Using real-time PCR, we confirmed that miR-21 was significantly overexpressed in human EOC tissues and cell lines. The overexpression of miR-21 correlated with histological differentiation, clinicopathological stage, and lymph node metastasis, and we showed that knockdown of miR-21 by an inhibitor caused a significant reduction in cell proliferation and decrease in cell migration and invasion abilities. Furthermore, we demon-strated that knockdown of miR-21 significantly increased the expression of PTEN, a known tumor suppressor in ovarian cancer. Collectively, our findings suggest miR-21 may be important in the initiation and progression of EOC as an oncomiR, likely through regulating PTEN.