Molecular biology of gynecological cancer (Review)

  • Authors:
    • Kenzo Sonoda
  • View Affiliations

  • Published online on: November 5, 2015     https://doi.org/10.3892/ol.2015.3862
  • Pages: 16-22
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer is a pathological condition in which the balance between cell growth and death is disordered. Various molecules have been reported to be involved in the oncogenic process of invasion, metastasis and resistance to treatment. An exponential growth in the collection of genomic and proteomic data in the past 20 years has provided major advances in understanding the molecular mechanisms of human cancer, which has been applied to diagnostic and treatment strategies. Targeted therapies have been developed and adopted, particularly for advanced, refractory or recurrent cancers, depending on individual molecular profiles. The aim of the present review is to provide a report of the current literature regarding the molecular biology of gynecological cancers.

Introduction

Cancer is a complex collection of diseases that arise due to genetic and epigenetic alterations that interfere with cellular growth and death (1). Specific molecular alterations that induce a normal cell to become malignant, defined as the ability to invade and metastaize from the primary lesion, have been revealed, but the spectrum of these alterations vary substantially among cancers. It is now recognized that metastasis is dependent on a balance of stimulating signals and inhibitory factors, which must be weighted in favor of the stimulating signals to result in metastasis (2). Stem cell studies have provided valuable information for cancer biology. As studies continue to understand the role of cancer stem cells, the resulting information is likely to lead to novel approaches to target cancer cells (3). Although conventional combinations of surgery, radiation and chemotherapy remain the mainstay of cancer treatment, it is challenging to treat advanced or recurrent cancers in this way. An exponential growth in the collection of genomic and proteomic data in the past 20 years has resulted in the development of various targeted therapies (4). In addition, recent advances in biochemical engineering should contribute to a major evolution in the diagnosis and treatment of human cancer (5). The initial sections of the present review examine the literature regarding the basic molecular mechanisms involved in the development of cancers and the expansion of the malignant phenotype. The molecular changes characteristic of gynecological cancers are outlined in section 5.

Oncogenesis

At least 3–6 changes are considered to be required to transform a cell (6). The majority of cancer cells are genetically unstable, and this leads to an accumulation of numerous secondary molecular alterations that play a role in the evolution of malignant characteristics, including immortality, invasion, metastasis and resistance to therapy (7). Epigenetics are hereditary changes that do not originate from DNA mutation (8). The primary mechanism of epigenetic alteration, which is regulated by a family of DNA methyltransferases, is methylation of cytosine residues that reside next to guanine (9). The majority of cancer types comprehensively reduce DNA methylation, a reduction that may contribute to genomic instability (10). Selective hypermethylation of cytosines in the promoter regions of tumor suppressor genes may lead to the inactivation of the genes, and this may result in carcinogenesis (11). Acetylation and methylation of the histone proteins that package and order the DNA into structural units, acts as another level of epigenetic regulation (11). These genetic and epigenetic alterations may become therapeutic targets against cancer.

Alterations in genes that stimulate cellular growth (oncogenes) are able to cause the malignant transformation of cells (12). Oncogenes are activated by several mechanisms, as follows: First, through the amplification of the oncogenes, which results in overexpression of the corresponding proteins; second, point mutations may overactivate oncogenes; and third, oncogenes may be translocated from one chromosome to another, affecting the promoter regions and resulting in overexpression of the gene (13). The third mechanism continually occurs in lymphomas and leukemias (14). Peptide growth factors, such as epidermal growth factor (EGF), platelet-derived growth factor, and fibroblast growth factor families, stimulate intracellular molecular pathways that lead to proliferation (1517). Growth factor receptors that exist on the cell membrane are composed of an extracellular domain, a transmembrane region, and a cytoplasmic tyrosine kinase domain (18). Following binding of a growth factor, the receptor undergoes a conformational change, resulting in the activation of the tyrosine kinase in the cytoplasmic domain. The tyrosine kinase phosphorylates tyrosine residues on the receptor itself and on downstream molecular targets in the cytoplasm (19).

G-protein coupled receptors (GPCRs) are cell membrane receptors that contain a 7-transmembrane α-helical fold, which transduces extracellular information into intracellular signal transduction pathways (20). GPCR signaling pathways are extremely complex and include the Ras superfamily guanosine triphosphatase (GTPase), one of the most frequently mutated oncogenes in humans (21). Harvey rat sarcoma viral oncogene homolog (H-RAS), Kirsten rat sarcoma viral oncogene homolog (K-RAS) and neuroblastoma rat sarcoma viral oncogene homolog are isoforms of the Ras protein and promote carcinogenesis when they are activated by mutations at codon 12, 13 or 61 (22). Although a high degree of similarity exists among the isoforms, K-RAS mutations are most frequently identified in cancer and each isoform demonstrates preferential association to particular cancer types (23). B-Raf proto-oncogene, serine/threonine kinase (BRAF) is a serine/threonine-specific protein kinase that interacts with Ras proteins in activating the MAP kinase pathway (24). Notably, the most common BRAF mutation, the V600E transversion, has been identified in numerous types of human cancer (25). Signals generated in the cell membrane and cytoplasm, following GPCR binding, result in the activation of nuclear transcription factors that are responsible for stimulating cell growth, including the FOS and JUN oncogenes that form the activator protein 1 transcription complex by dimerization (26). The MYC gene family is also involved in the development of human malignancies (27), and when inappropriately amplified or overexpressed, these transcription factors may act as oncogenes.

Loss of tumor suppressor gene function plays a role in tumorigenesis (28). Statistical analysis of cancer incidence has proposed the two-hit theory for the inactivation of tumor suppressor genes; mutations in each of the alleles are necessary for the loss of tumor suppressor gene function (7). However, subsequent interpretations of the analysis indicated that mutations in tumor suppressor genes are not absolutely recessive (7). Haploinsufficiency is a condition where only a single functional copy of a gene is produced (29). In a tumor suppressor gene, haploinsufficiency leads to the development of cancer (30). The phenotypic penetrance of tumor suppressor gene mutations may be affected, not only by the nature of the mutation itself, but also by other clinicopathological variables, including genetic background, tissue type and environmental factors (31). Certain tumor suppressor genes are inactivated by methylation of the promoter region of the gene (32). Examples include the retinoblastoma gene (RB1) in retinoblastoma (33), the Von Hippel-Lindau gene in renal carcinomas (34), the BRCA gene in breast cancer (35) and the APC gene in colorectal cancer (36). RB1 was the first tumor suppressor gene to be identified (37), and plays a pivotal role in the regulation of cell cycle progression. In the G1 phase of the cell cycle, the Rb protein, which is upregulated when RB1 is transcribed, binds to the E2F transcription factor and prevents cell cycle progression (38). When Rb is phosphorylated by cyclin and cyclin-dependent kinase (cdk) complexes, E2F is released and the cell cycle progresses. The RB1 gene mutation has been identified in retinoblastoma and certain types of sarcoma, but rarely in other types of cancer, unlike the tumor protein p53 (TP53) tumor suppressor gene, which is mutated in the majority of human cancers (39,40).

The p53 protein is a sensor of multiple forms of genetic toxicities and oncogenic stresses. Since the p53 protein represses growth and regulates the survival of stressed cells, it has been described as the ‘guardian of the genome’ (7). The clonal expansion of cells with the TP53 gene mutation may proceed to carcinogenesis (41). Numerous cancer cells possess missense mutations in exons 5–8 in one copy of the TP53 gene, which encode the DNA binding domains, and even though a deletion of the other copy of the TP53 gene does not occur, the mutant p53 protein is able to complex with the wild-type p53 protein and disturb the interaction between the wild-type p53 protein and DNA in a dominant negative fashion (42).

Although numerous tumor suppressor genes encode nuclear proteins, such as TP53 and RB1, certain intra-cytoplasmic tumor suppressor molecules have been identified, including: Phosphatase and tensin homolog (PTEN), which inhibits invasion and metastasis by modulating the cytoskeleton (43); a mutant version of the APC gene that is responsible for familial adenomatous polyposis syndrome (44); and transforming growth factor-β (TGF-β) protein, which is coded by the TGFB1 gene and inhibits the proliferation of normal epithelial cells (45). Additionally, the altered expression of microRNAs, which are single RNA strands of 21–23 nucleotides, has been detected in numerous cancers (46).

Families with inherited mutations in genes susceptible to cancer exhibit a high incidence of specific types of cancer (47). In addition, the age of cancer onset is younger and certain individuals experience multiple primary cancers (48,49). Mutations of tumor suppressor genes and DNA mismatch repair genes are usually involved in familial cancer syndromes, such as breast cancer 1, early onset (BRCA1) and breast cancer 2, early onset (BRCA2) in hereditary breast or ovarian cancer syndrome (50), APC in familial adenomatous polyposis syndrome (51), and MSH2, MLH1 and MSH6 in hereditary nonpolyposis colorectal cancer syndrome (HNPCC) (52).

Escape from cell death

The number of cells in normal tissues is strictly controlled by a balance between cell growth and death. Cell proliferation is regulated by the cell cycle, in which numerous molecules, are involved, including TP53, Rb, E2F, cyclins and cdks (53). Cancer cells lose cell cycle regulatory systems and are resistant to death and senescence (54). The three major cell death pathways are apoptosis, necrosis and autophagy (55). Apoptosis is an energy-dependent process in which DNA and proteins are cleaved by endonucleases and proteases, respectively (56). Apoptotic stimuli are delivered through an extrinsic or intrinsic pathway and may contribute to the prevention of cancer by eliminating transformed cells (57). Necrosis of cells instigates a strong immune response, and various anticancer agents stimulate a beneficial immune response by enhancing the necrosis of tumor cells (58). Autophagy is characterized by the degradation of cellular proteins and organelles in fused cytoplasmic autophagic vesicles and lysosomes (59). Cellular senescence is regulated by progressive shortening of telomeres (60). Cancer cells are capable of avoiding senescence by enhancing a ribonucleoprotein complex telomerase, and in gynecological malignancies high activity levels of telomerase are identified (61).

With regard to immune surveillance, cancer cells have developed mechanisms to evade immune responses and prolong survival. The receptor-binding cancer antigen (RCAS1), which is expressed on SiSo cells, may induce apoptosis in immune cells, including peripheral lymphocytes and natural killer cells (62). The involvement of RCAS1 in tumor cell escape from immune surveillance has been investigated using clinical specimens, as reviewed by Giaginis et al (63). In cervical cancer, the number of tumor cells expressing RCAS1 was significantly associated with the number of apoptotic lymphocytes at the primary site and in metastatic lymph nodes (64). In glioma and oral squamous cell, lung, breast, esophageal, gastric, biliary tract and colon cancers, increased numbers of apoptotic lymphocytes or decreased numbers of tumor-infiltrating lymphocytes have been reported (65). These observations suggest that RCAS1 plays a pivotal role in tumor cell evasion of immune surveillance. The signal transduction pathways that induce apoptosis following RCAS1 stimulation have been assessed; RCAS1 induced p38 MAPK phosphorylation (66), cytochrome c release and activation of caspase-3, but decreased cyclin D3 levels (67). However, RCAS1-induced apoptosis is strongly inhibited by the cysteine protease inhibitor benzyloxycarbonyl-Val-Ala-Asp fluoromethylketone (62).

Angiogenesis, invasion and metastasis

Angiogenesis is required for cancer cell proliferation. Various mediators of angiogenesis have been reported, including vascular endothelial growth factor (VEGF), interleukin-8 and matrix metalloproteinases (MMP) (68). Targeted therapy against angiogenesis using bevacizumab, a humanized VEGF-neutralizing monoclonal antibody, improved the median progression-free survival (PFS) rate in advanced ovarian cancer (69). Burger et al reported that the use of bevacizumab during and following carboplatin and paclitaxel chemotherapy prolonged the median progression-free survival time in patients with advanced epithelial ovarian cancer (69). In addition, debulking surgery following neoadjuvant chemotherapy, such as bevacizumab, did not increase the rate of post-operative complications (70).

A crucial first step of metastasis is invasion of cancer cells through the basement membrane of tissues. MMPs promote cancer cell invasion and endothelial cell migration (71) and were reported to be expressed at the invasive front of endometrial and ovarian cancer (72). Furthermore ovarian cancers that overexpress MMP-2 and MMP-9 demonstrate aggressive clinical characteristics (73,74). Adhesion molecules including focal adhesion kinase, integrins and E-cadherin may facilitate cancer progression. Sawada et al reported that α5-integrin upregulation was a molecular mechanism that led to a loss in E-cadherin, thereby promoting ovarian cancer cell metastasis (75). The propensity of various types of cancer to metastasize in specific organs was first proposed by Paget in 1889 (76). Paget's hypothesis stated that metastasis resulted from the dependence of the seed (cancer cell) on the soil (metastatic site), and was devised due to the non-random pattern observed with metastasis. Recent studies provide important information for this non-random pattern of metastasis. Breast cancers frequently metastasize to the lung, liver and bone marrow (77). Tumor cells also express high levels of chemokine (C-X-C motif) receptor 4 and CC chemokine receptor 7, and chemokine proteins chemokine (C-X-C motif) ligand 12 and chemokine (C-C motif) ligand 21 are detected at high levels in metastatic sites (78).

Several studies have suggested that RCAS1 may be involved in the aggressive characteristics of human malignancies, not only by helping tumor cells to evade immune surveillance, but also by inducing cancer stromal tissue remodeling (79). For example, in cervical cancer, RCAS1 expression levels were significantly correlated with those of MMP-1, an interstitial collagenase, and laminin-5, an extracellular matrix molecule (80), which have been reported to be involved in tumor invasion and metastasis (8182). Additionally, the number of stromal vimentin-positive cells was revealed to decrease in association with the RCAS1 expression level in cervical and ovarian cancer (79,84). The reduction in vimentin expression may result in tumor progression, since vimentin is involved in apoptosis (83) and the mechanical stability of stromal cells (8587). Furthermore, RCAS1 expression is significantly associated with VEGF expression and microvessel density in cervical cancer (88). RCAS1 is hypothesized to induce VEGF expression through the TGF-β and MAPK signaling pathways. Liby et al reported that a blockade of protein kinase B (Akt)-3 resulted in smaller and less vascularized tumors with a downregulation of RCAS1 and VEGF expression in a xenograft mouse model (89). MMPs and laminin also play an important role in angiogenesis via regulation of extracellular matrix degradation and remodeling (90).

Gynecological malignancies

Cervical cancer

Worldwide, cervical cancer was the fourth most commonly diagnosed malignancy in women in 2012, with an estimated 527,600 novel cases and 265,700 fatalities (91). Overall, ~85% of novel cases and mortalities occurred in developing countries. Human papilloma virus (HPV) has been reported to play a pivotal role in the carcinogenesis of cervical cancer (92). HPV DNA is composed of 7,800 nucleotides that include early and late open reading frames (ORFs) (93). Early ORFs encode 7 proteins, termed E1–7, that are involved in viral replication and host cell transformation (94). Late ORFs encode structural proteins of the virion, such as L1 and 2. Cellular transformation may be associated with the integration of HPV DNA into the host genome, for example E6 and 7 bind to and inactivate p53 and Rb, respectively, resulting in transformation. This observation suggests that the oncogenic potential of HPV is associated with the binding affinities of E6 and 7 (95,96).

The majority of HPV infections spontaneously regress, and only a small proportion of HPV-infected individuals develop cervical cancer (97). Additive molecular alterations to HPV infections are considered to be required for the progression to cervical cancer, but these changes are not yet fully understood. Comparative genomic hybridization analysis has revealed the addition of chromosomes 1q and 3q and the loss of chromosomes 2q, 3p and 11q in cervical cancer (97). The fragile histidine triad gene (FHIT) is located in chromosome 3p and FHIT expression is frequently reduced in cervical cancer (98). Since allelic losses and homozygous deletions, as well as loss of heterozygosity, are frequently detected in microsatellites in FHIT, alterations and inactivation of the FHIT gene are presumed to contribute to and accelerate cervical carcinogenesis, for example decreased expression of FHIT and overexpression of p16 and c-myc are considered to be early events in cervical cancer (99). Mutations in the K-RAS or H-RAS genes are suggested to be a late event for cervical carcinogenesis (100). RCAS1 expression has been immunohistochemically studied during the carcinogenesis of cervical cancer and was not detected in dysplasias; however, 20% of carcinomas in situ, 16% of microinvasive carcinomas, and 82% of invasive carcinomas stained for the expression of RCAS1 (101). Therefore, RCAS1 expression may be associated with tumor progression and invasion in the squamous cell epithelium of the cervix.

Endometrial cancer

Endometrial cancer is the most commonly diagnosed gynecological malignancy in developed countries (102). In the USA, a projected 54,870 patients are expected to be diagnosed and 10,170 cases are expected to be fatal in 2015 (103). Between 2006 and 2010, the incidence of novel cases of endometrial cancer increased by 1.5% per year among females aged <50 years and 2.6% per year among females aged ≥50 years. In the same period, mortality from endometrial cancer increased by 1.5% per year among females aged <50 years, but were stable among females aged ≥50 years. The majority of endometrial cancers are sporadic, ~5% of cases are considered to be hereditary and caused by DNA mismatch repair gene mutations (104). Endometrial cancer is the second most commonly diagnosed malignancy in females with HNPCC (105).

Endometrial cancers are classified as Type I or II, according to clinicopathological variables (106). Type I endometrial cancer arises from preneoplastic lesion hyperplasia that has undergone unchecked estrogenic stimulation (107). During menopause, estrogens are produced from androgens secreted from adrenal glands and non-specialized ovarian stroma. Niwa et al reported that estrone was the most effective estrogen to promote endometrial carcinogenesis in a mouse model (108). Almost 80% of Type I endometrioid adenocarcinomas exhibited a loss of PTEN expression (109), and mutations of K-RAS and β-catenin are frequently detected (110). Microsatellite instability (MSI) is a significant genetic alteration demonstrated in almost 45% of endometrial cancer lesions (111) that results from impaired DNA mismatch repair. Cells without normally functioning DNA mismatch repair cannot correct errors, including single base mismatches, insertions and deletions during DNA replication. Mismatch repair gene MLH1 deficiency, caused by epigenetic hypermethylation in its promoter region, is most frequently observed in sporadic endometrial adenocarcinomas. Loss of PTEN expression, K-RAS mutation and MSI are suggested to be early events in endometrial carcinogenesis. Type II carcinomas develop from atrophic endometrium and are frequently serous or clear cell adenocarcinomas. TP53 mutations occur in ~90% of serous adenocarcinomas and are almost always associated with aneuploidy (112). p16 inactivation, HER2 overexpression, and reduced E-cadherin expression are observed in ~45, 70, and 80% of cases, respectively (113,114).

RCAS1 expression was investigated during the carcinogenesis of endometrial cancer, and was significantly increased in endometrial adenocarcinoma compared with normal cells or hyperplasias, suggesting that RCAS1 expression may be associated with oncogenic transformation (115). RCAS1 expression was revealed to be inversely correlated with the prognosis of patients with endometrial cancer (116).

Ovarian cancer

The majority of ovarian cancers are sporadic and arise from an accumulation of genetic damage (117). Ovarian cancers are heterogeneous regarding histopathology and malignant potential; therefore, the characteristic patterns of molecular signature are also varied (118). Serous borderline tumor and low-grade serous adenocarcinoma are frequently characterized by mutations in K-RAS and BRAF (119). Sporadic high-grade serous adenocarcinoma is usually advanced at diagnosis and the prognosis of patients is poor. Ovarian cancer has frequent mutations in TP53 and occasional overexpression of HER-2/neu, AKT2 and MYC (120,121). Endometrioid and clear cell adenocarcinomas have been suggested to associate with endometriosis and possess frequent mutations of PTEN and PIK3CA (122). Mucinous adenocarcinoma is usually diagnosed at an early stage of disease and characterized by mutations in K-RAS (123). In total, ~10% of ovarian cancers are attributable to inherited mutations of cancer susceptibility genes, including BRCA1 and BRCA2 (124), and high-grade serous adenocarcinoma is a predominant histological type in patients with inherited BRCA mutations. BRCA1 and BRCA2 are located in chromosome 17q and 13q, respectively (125). Since the BRCA1 and BRCA2 proteins complex with Rad51 recombinase and other molecules that are involved in the repair of DNA double strand breaks by homologous recombination, BRCA1 and BRCA2 are classified as tumor suppressor genes (126). The life-time risk of ovarian cancer ranges between 20–40% and 10–20% in BRCA1 and BRCA2 carriers, respectively, and the median age of patients is 40–50 years old (127129). Poly(adenosine diphosphate-ribose) polymerase (PARP) is involved in the repair of DNA single strand breaks by base excision. The efficacy of a PARP inhibitor was investigated in ovarian cancer patients with BRCA mutations (130). Randomized phase II studies of platinum-sensitive recurrent high-grade serous cancers (both germline BRCA and sporadic) showed that patients treated with a PARP inhibitor exhibited a significant improvement in PFS when compared with patients who were administered a placebo (131).

Lysophosphatidic acid, heparin-binding EGF (HB-EGF) and amphiregulin, have been reported to play pivotal roles in proliferation and dissemination of ovarian cancer (132,133). Yagi et al reported that inhibitory agents against HB-EGF, including CRM197, were possible chemotherapeutic and chemosensitizing agents for ovarian cancer (134). The clinical application of novel treatments which target these molecules is expected in the future.

Conclusion

Since it is challenging to treat advanced or recurrent cancers using conventional treatments, the development of novel and highly specific targets for therapy is required. An exponential growth in the collection of genomic and proteomic data in the past 20 years has provided major advances in understanding the molecular mechanisms of human cancer. This progress has resulted in the development of targeted therapies tailored to an individual molecular profile. Recent advances in biochemical engineering should contribute to major evolution in diagnosis and treatment of human cancer.

Acknowledgements

This review was supported in part by a Grant-in-Aid for Scientific Research from the Japan Society for the Promotion of Science (grant no. 24592519).

Glossary

Abbreviations

Abbreviations:

EGF

epidermal growth factor

GPCR

G-protein coupled receptor

RB1

retinoblastoma gene

cdk

cyclin-dependent kinase

TGF-β

transforming growth factor-β

HNPCC

hereditary nonpolyposis colorectal cancer syndrome

RCAS1

receptor-binding cancer antigen expressed on SiSo cells

VEGF

vascular endothelial growth factor

MMP

matrix metalloproteinase

PFS

progression-free survival

HPV

human papilloma virus

ORF

open reading frame

FHIT

fragile histidine triad gene

MSI

microsatellite instability

PARP

poly (ADP-ribose) polymerase

HB-EGF

heparin-binding EGF

References

1 

Hoeijmakers JH: DNA damage, aging, and cancer. N Engl J Med. 361:1475–1485. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Entschladen F, Drell TL IV, Lang K, Joseph J and Zaenker KS: Tumour-cell migration, invasion, and metastasis: Navigation by neurotransmitters. Lancet Oncol. 5:254–258. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Meacham CE and Morrison SJ: Tumour heterogeneity and cancer cell plasticity. Nature. 501:328–337. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Mok TS: Personalized medicine in lung cancer: What we need to know. Nat Rev Clin Oncol. 8:661–668. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Diou O, Tsapis N and Fattal E: Targeted nanotheranostics for personalized cancer therapy. Expert Opin Drug Deliv. 9:1475–1487. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Croce CM: Oncogenes and cancer. N Engl J Med. 358:502–511. 2008. View Article : Google Scholar : PubMed/NCBI

7 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Esteller M: Epigenetics in cancer. N Engl J Med. 358:1148–1159. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Berdasco M and Esteller M: Aberrant epigenetic landscape in cancer: How cellular identity goes awry. Dev Cell. 19:698–711. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Esteller M: Cancer epigenomics: DNA methylomes and histone-modification maps. Nat Rev Genet. 8:286–298. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Jones PA and Baylin SB: The epigenomics of cancer. Cell. 128:683–692. 2007. View Article : Google Scholar : PubMed/NCBI

12 

Doroshow JH and Kummar S: Translational research in oncology-10 years of progress and future prospects. Nat Rev Clin Oncol. 11:649–662. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Negrini S, Gorgoulis VG and Halazonetis TD: Genomic instability - an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 11:220–228. 2010. View Article : Google Scholar : PubMed/NCBI

14 

Tomita N: BCL2 and MYC dual-hit lymphoma/leukemia. J Clin Exp Hematop. 51:7–12. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Maire CL and Ligon KL: Molecular pathologic diagnosis of epidermal growth factor receptor. Neuro Oncol. 16:viii1–6. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Demoulin JB and Essaghir A: PDGF receptor signaling networks in normal and cancer cells. Cytokine Growth Factor Rev. 25:273–283. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Fearon AE, Gould CR and Grose RP: FGFR signaling in women's cancers. Int J Biochem Cell Biol. 45:2832–2842. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Normanno N, De Luca A, Bianco C, Strizzi L, Mancino M, Maiello MR, Carotenuto A, De Feo G, Caponigro F and Salomon DS: Epidermal growth factor receptor (EGFR) signaling in cancer. Gene. 366:2–16. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Bishayee A, Beguinot L and Bishayee S: Phosphorylation of tyrosine 992, 1068, and 1086 is required for conformational change of the human epidermal growth factor receptor c-terminal tail. Mol Biol Cell. 10:525–536. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Heng BC, Aubel D and Fussenegger M: An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv. 31:1676–1694. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Shaw RJ and Cantley LC: Ras, PI(3)K and mTOR signaling controls tumour cell growth. Nature. 441:424–430. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Prior IA, Lewis PD and Mattos C: A comprehensive survey of Ras mutations in cancer. Cancer Res. 72:2457–2467. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Fernández-Medarde A and Santos E: Ras in cancer and developmental diseases. Genes Cancer. 2:344–358. 2011. View Article : Google Scholar : PubMed/NCBI

24 

Ho CL, Kurman RJ, Dehari R, Wang TL and Shih IeM: Mutations of BRAF and KRAS precede the development of ovarian serous borderline tumors. Cancer Res. 64:6915–6918. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Cantwell-Dorris ER, O'Leary JJ and Sheils OM: BRAFV600E: Implications for carcinogenesis and molecular therapy. Mol Cancer Ther. 10:385–394. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Eferl R and Wagner EF: AP-1: A double-edged sword in tumorigenesis. Nat Rev Cancer. 3:859–868. 2003. View Article : Google Scholar : PubMed/NCBI

27 

Li Y, Casey SC and Felsher DW: Inactivation of MYC reverses tumorigenesis. J Intern Med. 276:52–60. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Payne SR and Kemp CJ: Tumor suppressor genetics. Carcinogenesis. 26:2031–2045. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Berger AH and Pandolfi PP: Haplo-insufficiency: A driving force in cancer. J Pathol. 223:137–146. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Smith U: PTEN - linking metabolism, cell growth, and cancer. N Engl J Med. 367:1061–1063. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Chaudru V, Chompret A, Bressac-de Paillerets B, Spatz A, Avril MF and Demenais F: Influence of genes, nevi, and sun sensitivity on melanoma risk in a family sample unselected by family history and in melanoma-prone families. J Natl Cancer Inst. 96:785–795. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Sugimura T and Ushijima T: Genetic and epigenetic alterations in carcinogenesis. Mutat Res. 462:235–246. 2000. View Article : Google Scholar : PubMed/NCBI

33 

Greger V, Debus N, Lohmann D, Höpping W, Passarge E and Horsthemke B: Frequency and parental origin of hypermethylated RB1 alleles in retinoblastoma. Hum Genet. 94:491–496. 1994. View Article : Google Scholar : PubMed/NCBI

34 

Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, Samid D, Duan DS, Gnarra JR, Linehan WM, et al: Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci USA. 91:9700–9704. 1994. View Article : Google Scholar : PubMed/NCBI

35 

Dobrovic A and Simpfendorfer D: Methylation of the BRCA1 gene in sporadic breast cancer. Cancer Res. 57:3347–3350. 1997.PubMed/NCBI

36 

Arnold CN, Goel A, Niedzwiecki D, Dowell JM, Wasserman L, Compton C, Mayer RJ, Bertagnolli MM and Boland CR: APC promoter hypermethylation contributes to the loss of APC expression in colorectal cancers with allelic loss on 5q. Cancer Biol Ther. 3:960–964. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Madhavan J, Ganesh A and Kumaramanickavel G: Retinoblastoma: From disease to discovery. Ophthalmic Res. 40:221–226. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Guimaraes DP and Hainaut P: TP53: A key gene in human cancer. Biochimie. 84:83–93. 2002. View Article : Google Scholar : PubMed/NCBI

39 

Lipinski MM and Jacks T: The retinoblastoma gene family in differentiation and development. Oncogene. 18:7873–7882. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Soussi T: The history of p53. A perfect example of the drawbacks of scientific paradigms. EMBO Rep. 11:822–826. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Ghebranious N and Donehower LA: Mouse models in tumor suppression. Oncogene. 17:3385–3400. 1998. View Article : Google Scholar : PubMed/NCBI

42 

Walerych D, Napoli M, Collavin L and Del Sal G: The rebel angel: Mutant p53 as the driving oncogene in breast cancer. Carcinogenesis. 33:2007–2017. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Chalhoub N and Baker SJ: PTEN and the PI3-kinase pathway in cancer. Annu Rev Pathol. 4:127–150. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Jaiswal AS and Narayan S: A novel function of adenomatous polyposis coli (APC) in regulating DNA repair. Cancer Lett. 271:272–280. 2008. View Article : Google Scholar : PubMed/NCBI

45 

Tian M and Schiemann WP: The TGF-beta paradox in human cancer: An update. Future Oncol. 5:259–271. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Kong YW, Ferland-McCollough D, Jackson TJ and Bushell M: microRNAs in cancer management. Lancet Oncol. 13:e249–258. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Lu KH: Hereditary gynecologic cancers: Differential diagnosis, surveillance, management and surgical prophylaxis. Fam Cancer. 7:53–58. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Perea J, Rodríguez Y, Rueda D, Marín JC, Díaz-Tasende J, Álvaro E, Alegre C, Osorio I, Colina F, Lomas M, et al: Early-onset colorectal cancer is an easy and effective tool to identify retrospectively Lynch syndrome. Ann Surg Oncol. 18:3285–3291. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Nieuwenhuis MH, Kets CM, Murphy-Ryan M, Yntema HG, Evans DG, Colas C, Møller P, Hes FJ, Hodgson SV, Olderode-Berends MJ, et al: Cancer risk and genotype-phenotype correlations in PTEN hamartoma tumor syndrome. Fam Cancer. 13:57–63. 2014. View Article : Google Scholar : PubMed/NCBI

50 

Kobayashi H, Ohno S, Sasaki Y and Matsuura M: Hereditary breast and ovarian cancer susceptibility genes (review). Oncol Rep. 30:1019–1029. 2013.PubMed/NCBI

51 

Rozen P and Macrae F: Familial adenomatous polyposis: The practical applications of clinical and molecular screening. Fam Cancer. 5:227–235. 2006. View Article : Google Scholar : PubMed/NCBI

52 

Vasen HF, Möslein G, Alonso A, et al: Guidelines for the clinical management of Lynch syndrome (hereditary non-polyposis cancer). J Med Genet. 44:353–362. 2007. View Article : Google Scholar : PubMed/NCBI

53 

Harris SL and Levine AJ: The p53 pathway: Positive and negative feedback loops. Oncogene. 24:2899–2908. 2005. View Article : Google Scholar : PubMed/NCBI

54 

Collado M and Serrano M: Senescence in tumours: Evidence from mice and humans. Nat Rev Cancer. 10:51–57. 2010. View Article : Google Scholar : PubMed/NCBI

55 

Su Z, Yang Z, Xu Y, Chen Y and Yu Q: Apoptosis, autophagy, necroptosis, and cancer metastasis. Mol Cancer. 14:482015. View Article : Google Scholar : PubMed/NCBI

56 

Hensley P, Mishra M and Kyprianou N: Targeting caspases in cancer therapeutics. Biol Chem. 394:831–843. 2013. View Article : Google Scholar : PubMed/NCBI

57 

Lowe SW, Cepero E and Evan G: Intrinsic tumour suppression. Nature. 432:307–315. 2004. View Article : Google Scholar : PubMed/NCBI

58 

Van den Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H and Vandenabeele P: Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 15:135–147. 2014. View Article : Google Scholar : PubMed/NCBI

59 

Chaabane W, User SD, El-Gazzah M, Jaksik R, Sajjadi E, Rzeszowska-Wolny J and Los MJ: Autophagy, apoptosis, mitoptosis and necrosis: Interdependence between those pathways and effects on cancer. Arch Immunol Ther Exp (Warsz). 61:43–58. 2013. View Article : Google Scholar : PubMed/NCBI

60 

van Deursen JM: The role of senescent cells in ageing. Nature. 509:439–446. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Sakamoto M, Toyoizumi T, Kikuchi Y, Okamoto A, Nakayama H, Aoki D, Yamamoto K, Hata H, Sugishita T and Tenjin Y: Telomerase activity in gynecological tumors. Oncol Rep. 7:1003–1009. 2000.PubMed/NCBI

62 

Nakashima M, Sonoda K and Watanabe T: Inhibition of cell growth and induction of apoptotic cell death by the human tumor-associated antigen RCAS1. Nat Med. 5:938–942. 1999. View Article : Google Scholar : PubMed/NCBI

63 

Giaginis C, Giagini A and Theocharis S: Receptor-binding cancer antigen expressed on SiSo cells (RCAS1): A novel biomarker in the diagnosis and prognosis of human neoplasia. Histol Histopathol. 24:761–776. 2009.PubMed/NCBI

64 

Sonoda K, Miyamoto S, Hirakawa T, Yagi H, Yotsumoto F, Nakashima M, Watanabe T and Nakano H: Association between RCAS1 expression and microenvironmental immune cell death in uterine cervical cancer. Gynecol Oncol. 97:772–779. 2005. View Article : Google Scholar : PubMed/NCBI

65 

Sonoda K: RCAS1 is a promising therapeutic target against cancer: Its multifunctional bioactivities and clinical significance. Expert Rev Obstet Gynecol. 7:261–267. 2012. View Article : Google Scholar

66 

Sonoda K, Miyamoto S, Nakashima M and Wake N: The biological role of unique molecule RCAS1: A bioactive marker that induces connective tissue remodeling and lymphocyte apoptosis. Front Biosci. 13:1106–1116. 2008. View Article : Google Scholar : PubMed/NCBI

67 

Nishinakagawa T, Fujii S, Nozaki T, Maeda T, Machida K, Enjoji M and Nakashima M: Analysis of cell cycle arrest and apoptosis induced by RCAS1. Int J Mol Med. 25:717–722. 2010.PubMed/NCBI

68 

Baeriswyl V and Christofori G: The angiogenic switch in carcinogenesis. Semin Cancer Biol. 19:329–337. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Burger RA, Brady MF, Bookman MA, et al: Gynecologic Oncology Group: Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med. 365:2473–2483. 2011. View Article : Google Scholar : PubMed/NCBI

70 

Chéreau E, Lambaudie E and Houvenaeghel G: Morbidity of surgery after neoadjuvant chemotherapy including bevacizumab for advanced ovarian cancer. Int J Gynecol Cancer. 23:1326–1330. 2013. View Article : Google Scholar : PubMed/NCBI

71 

Hua H, Li M, Luo T, Yin Y and Jiang Y: Matrix metalloproteinases in tumorigenesis: An evolving paradigm. Cell Mol Life Sci. 68:3853–3868. 2011. View Article : Google Scholar : PubMed/NCBI

72 

Planagumà J, Liljeström M, Alameda F, Bützow R, Virtanen I, Reventós J and Hukkanen M: Matrix metalloproteinase-2 and matrix metalloproteinase-9 codistribute with transcription factors RUNX1/AML1 and ETV5/ERM at the invasive front of endometrial and ovarian carcinoma. Hum Pathol. 42:57–67. 2011. View Article : Google Scholar : PubMed/NCBI

73 

Yan L, Lin B, Gao L, Gao S, Liu C, Wang C, Wang Y, Zhang S and Iwamori M: Lewis (y) antigen overexpression increases the expression of MMP-2 and MMP-9 and invasion of human ovarian cancer cells. Int J Mol Sci. 11:4441–4452. 2010. View Article : Google Scholar : PubMed/NCBI

74 

Kamat AA, Fletcher M, Gruman LM, Mueller P, Lopez A, Landen CN Jr, Han L, Gershenson DM and Sood AK: The clinical relevance of stromal matrix metalloproteinase expression in ovarian cancer. Clin Cancer Res. 12:1707–1714. 2006. View Article : Google Scholar : PubMed/NCBI

75 

Sawada K, Mitra AK, Radjabi AR, Bhaskar V, Kistner EO, Tretiakova M, Jagadeeswaran S, Montag A, Becker A, Kenny HA, et al: Loss of E-cadherin promotes ovarian cancer metastasis via alpha 5-integrin, which is a therapeutic target. Cancer Res. 68:2329–2339. 2008. View Article : Google Scholar : PubMed/NCBI

76 

Paget S: The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 8:98–101. 1989.PubMed/NCBI

77 

Zhang Y, Ma B and Fan Q: Mechanisms of breast cancer bone metastasis. Cancer Lett. 292:1–7. 2010. View Article : Google Scholar : PubMed/NCBI

78 

Mukherjee D and Zhao J: The role of chemokine receptor CXCR4 in breast cancer metastasis. Am J Cancer Res. 3:46–57. 2013.PubMed/NCBI

79 

Sonoda K: Novel therapeutic strategies to target RCAS1, which induces apoptosis via ectodomain shedding. Histol Histopathol. 26:1475–1486. 2011.PubMed/NCBI

80 

Sonoda K, Miyamoto S, Hirakawa T, Yagi H, Yotsumoto F, Nakashima M, Watanabe T and Nakano H: Invasive potency related to RCAS1 expression in uterine cervical cancer. Gynecol Oncol. 99:189–198. 2005. View Article : Google Scholar : PubMed/NCBI

81 

Skyldberg B, Salo S, Eriksson E, Aspenblad U, Moberger B, Tryggvason K and Auer G: Laminin-5 as a marker of invasiveness in cervical lesions. J Natl Cancer Inst. 91:1882–1887. 1999. View Article : Google Scholar : PubMed/NCBI

82 

Brinkerhoff CE, Rutter JL and Benbow U: Interstitial collagenases as markers of tumor progression. Clin Cancer Res. 6:4823–4830. 2000.PubMed/NCBI

83 

Przybylo JA and Radisky DC: Matrix metalloproteinase-induced epithelial-mesenchymal transition: Tumor progression at Snail's pace. Int J Biochem Cell Biol. 39:1082–1088. 2007. View Article : Google Scholar : PubMed/NCBI

84 

Sonoda K, Miyamoto S, Kobayashi H, Ogawa S, Okugawa K, Taniguchi S and Wake N: The level of RCAS1 expression is inversely correlated with the number of vimentin-positive stromal cells in epithelial ovarian cancer. Int J Gynecol Cancer. 19:838–843. 2009. View Article : Google Scholar : PubMed/NCBI

85 

Byun Y, Chen F, Chang R, Trivedi M, Green KJ and Cryns VL: Caspase cleavage of vimentin disrupts intermediate filaments and promotes apoptosis. Cell Death Differ. 8:443–450. 2001. View Article : Google Scholar : PubMed/NCBI

86 

Eckes B, Dogic D, Colucci-Guyon E, Wang N, Maniotis A, Ingber D, Merckling A, Langa F, Aumailley M, Delouvée A, et al: Impaired mechanical stability, migration and contractile capacity in vimentin-deficient fibroblasts. J Cell Sci. 111:1897–1907. 1998.PubMed/NCBI

87 

Nieminen M, Henttinen T, Merinen M, Marttila-Ichihara F, Eriksson JE and Jalkanen S: Vimentin function in lymphocyte adhesion and transcellular migration. Nat Cell Biol. 8:156–162. 2006. View Article : Google Scholar : PubMed/NCBI

88 

Sonoda K, Miyamoto S, Yamazaki A, Kobayashi H, Nakashima M, Mekada E and Wake N: Biologic significance of receptor-binding cancer antigen expressed on SiSo cells (RCAS1) as a pivotal regulator of tumor growth through angiogenesis in human uterine cancer. Cancer. 110:1979–1990. 2007. View Article : Google Scholar : PubMed/NCBI

89 

Liby TA, Spyropoulos P, Buff Lindner H, Eldridge J, Beeson C, Hsu T and Muise-Helmericks RC: Akt3 controls vascular endothelial growth factor secretion and angiogenesis in ovarian cancer cells. Int J Cancer. 130:532–543. 2012. View Article : Google Scholar : PubMed/NCBI

90 

Davis GE and Senger DR: Endothelial extracellular matrix: Biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ Res. 97:1093–1107. 2005. View Article : Google Scholar : PubMed/NCBI

91 

American Cancer Society: Global Cancer Facts & Figures (3rd). Atlanta, GA: American Cancer Society. 34–36. 2015.

92 

Schiffman M, Castle PE, Jeronimo J, et al: Human papillomavirus and cervical cancer. Lancet. 370:890–907. 2007. View Article : Google Scholar : PubMed/NCBI

93 

Burk RD, Chen Z and Van Doorslaer K: Human papillomaviruses: Genetic basis of carcinogenicity. Public Health Genomics. 12:281–290. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Moody CA and Laimins LA: Human papillomavirus oncoproteins: Pathways to transformation. Nat Rev Cancer. 10:550–560. 2010. View Article : Google Scholar : PubMed/NCBI

95 

Buck CB, Day PM and Trus BL: The papillomavirus major capsid protein L1. Virology. 445:169–174. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Mammas IN, Sourvinos G, Giannoudis A and Spandidos DA: Human papilloma virus (HPV) and host cellular interactions. Pathol Oncol Res. 14:345–354. 2008. View Article : Google Scholar : PubMed/NCBI

97 

Huang KF, Lee WY, Huang SC, Lin YS, Kang CY, Liou CP and Tzeng CC: Chromosomal gain of 3q and loss of 11q often associated with nodal metastasis in early stage cervical squamous cell carcinoma. J Formos Med Assoc. 106:894–902. 2007. View Article : Google Scholar : PubMed/NCBI

98 

Giarnieri E, Zanesi N, Bottoni A, Alderisio M, Lukic A, Vecchione A, Ziparo V, Croce CM and Mancini R: Oncosuppressor proteins of fragile sites are reduced in cervical cancer. Cancer Lett. 289:40–45. 2010. View Article : Google Scholar : PubMed/NCBI

99 

Samir R, Asplund A, Tot T, Pekar G and Hellberg D: High-risk HPV infection and CIN grade correlates to the expression of c-myc, CD4+, FHIT, E-cadherin, Ki-67, and p16INK4a. J Low Genit Tract Dis. 15:280–286. 2011. View Article : Google Scholar : PubMed/NCBI

100 

Spandidos DA, Dokianakis DN, Kallergi G and Aggelakis E: Molecular basis of gynecological cancer. Ann N Y Acad Sci. 900:56–64. 2000. View Article : Google Scholar : PubMed/NCBI

101 

Sonoda K, Kaku T, Kamura T, Nakashima M, Watanabe T and Nakano H: Tumor-associated antigen 22-1-1 expression in the uterine cervical squamous neoplasia. Clin Cancer Res. 4:1517–1520. 1998.PubMed/NCBI

102 

Wright JD, Barrena Medel NI, Sehouli J, Fujiwara K and Herzog TJ: Contemporary management of endometrial cancer. Lancet. 379:1352–1360. 2012. View Article : Google Scholar : PubMed/NCBI

103 

American Cancer Society: Cancer facts & figures 2015. Atlanta, GA: American Cancer Society. 24–25. 2015.

104 

Sorosky JI: Endometrial cancer. Obstet Gynecol. 120:383–397. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Koornstra JJ, Mourits MJ, Sijmons RH, Leliveld AM, Hollema H and Kleibeuker JH: Management of extracolonic tumours in patients with Lynch syndrome. Lancet Oncol. 10:400–408. 2009. View Article : Google Scholar : PubMed/NCBI

106 

Matias-Guiu X and Prat J: Molecular pathology of endometrial carcinoma. Histopathology. 62:111–123. 2013. View Article : Google Scholar : PubMed/NCBI

107 

Banno K, Yanokura M, Iida M, Masuda K and Aoki D: Carcinogenic mechanisms of endometrial cancer: Involvement of genetics and epigenetics. J Obstet Gynaecol Res. 40:1957–1967. 2014. View Article : Google Scholar : PubMed/NCBI

108 

Niwa K, Murase T, Furui T, Morishita S, Mori H, Tanaka T, Mori H and Tamaya T: Enhancing effects of estrogens on endometrial carcinogenesis initiated by N-methyl-N-nitrosourea in ICR mice. Jpn J Cancer Res. 84:951–955. 1993. View Article : Google Scholar : PubMed/NCBI

109 

Ohgami T and Kato K: Pathogenesis of endometrial cancer. Current Approaches to Endometrial Cancer. Sakuragi N and Silverberg SG: (London). Future Medicine. 18–32. 2014.

110 

Yeramian A, Moreno-Bueno G, Dolcet X, Catasus L, Abal M, Colas E, Reventos J, Palacios J, Prat J and Matias-Guiu X: Endometrial carcinoma: Molecular alterations involved in tumor development and progression. Oncogene. 32:403–413. 2013. View Article : Google Scholar : PubMed/NCBI

111 

MacDonald ND, Salvesen HB, Ryan A, Iversen OE, Akslen LA and Jacobs IJ: Frequency and prognostic impact of microsatellite instability in a large population-based study of endometrial carcinomas. Cancer Res. 60:1750–1752. 2000.PubMed/NCBI

112 

Hecht JL and Mutter GL: Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol. 24:4783–4791. 2006. View Article : Google Scholar : PubMed/NCBI

113 

Halperin R, Zehavi S, Habler L, Hadas E, Bukovsky I and Schneider D: Comparative immunohistochemical study of endometrioid and serous papillary carcinoma of endometrium. Eur J Gynaecol Oncol. 22:122–126. 2001.PubMed/NCBI

114 

Holcomb K, Delatorre R, Pedemonte B, McLeod C, Anderson L and Chambers J: E-cadherin expression in endometrioid, papillary serous, and clear cell carcinoma of the endometrium. Obstet Gynecol. 100:1290–1295. 2002. View Article : Google Scholar : PubMed/NCBI

115 

Sonoda K, Kaku T, Hirakawa T, Kobayashi H, Amada S, Sakai K, Nakashima M, Watanabe T and Nakano H: The clinical significance of tumor-associated antigen RCAS1 expression in the normal, hyperplastic, and malignant uterine endometrium. Gynecol Oncol. 79:424–429. 2000. View Article : Google Scholar : PubMed/NCBI

116 

Sonoda K, Miyamoto S, Hirakawa T, Kaku T, Nakashima M, Watanabe T, Akazawa K, Fujita T and Nakano H: Association between RCAS1 expression and clinical outcome in uterine endometrial cancer. Br J Cancer. 89:546–551. 2003. View Article : Google Scholar : PubMed/NCBI

117 

Landen CN Jr, Birrer MJ and Sood AK: Early events in the pathogenesis of epithelial ovarian cancer. J Clin Oncol. 26:995–1005. 2008. View Article : Google Scholar : PubMed/NCBI

118 

Gui T, Cao D, Yang J and Shen K: Tumor heterogeneity has important consequences for personalized medicine in ovarian cancer. Histol Histopathol. 30:173–181. 2015.PubMed/NCBI

119 

Mayr D, Hirschmann A, Löhrs U and Diebold J: KRAS and BRAF mutations in ovarian tumors: A comprehensive study of invasive carcinomas, borderline tumors and extraovarian implants. Gynecol Oncol. 103:883–887. 2006. View Article : Google Scholar : PubMed/NCBI

120 

O'Neill CJ, Deavers MT, Malpica A, Foster H and McCluggage WG: An immunohistochemical comparison between low-grade and high-grade ovarian serous carcinomas: Significantly higher expression of p53, MIB1, BCL2, HER-2/neu, and C-KIT in high-grade neoplasms. Am J Surg Pathol. 29:1034–1041. 2005.PubMed/NCBI

121 

Nowee ME, Snijders AM, Rockx DA, de Wit RM, Kosma VM, Hämäläinen K, Schouten JP, Verheijen RH, van Diest PJ, Albertson DG and Dorsman JC: DNA profiling of primary serous ovarian and fallopian tube carcinomas with array comparative genomic hybridization and multiplex ligation-dependent probe amplification. J Pathol. 213:46–55. 2007. View Article : Google Scholar : PubMed/NCBI

122 

Wiegand KC, Hennessy BT, Leung S, Wang Y, Ju Z, McGahren M, Kalloger SE, Finlayson S, Stemke-Hale K, Lu Y, et al: A functional proteogenomic analysis of endometrioid and clear cell carcinomas using reverse phase protein array and mutation analysis: Protein expression is histotype-specific and loss of ARID1A/BAF250a is associated with AKT phosphorylation. BMC Cancer. 14:1202014. View Article : Google Scholar : PubMed/NCBI

123 

Naik JD, Seligmann J and Perren TJ: Mucinous tumours of the ovary. J Clin Pathol. 65:580–584. 2012. View Article : Google Scholar : PubMed/NCBI

124 

George SH and Shaw P: BRCA and early events in the development of serous ovarian cancer. Front Oncol. 4:52014. View Article : Google Scholar : PubMed/NCBI

125 

Powell SN and Kachnic LA: Roles of BRCA1 and BRCA2 in homologous recombination, DNA replication fidelity and the cellular response to ionizing radiation. Oncogene. 22:5784–5791. 2003. View Article : Google Scholar : PubMed/NCBI

126 

Venkitaraman AR: Linking the cellular functions of BRCA genes to cancer pathogenesis and treatment. Annu Rev Pathol. 4:461–487. 2009. View Article : Google Scholar : PubMed/NCBI

127 

Risch HA, McLaughlin JR, Cole DE, Rosen B, Bradley L, Kwan E, Jack E, Vesprini DJ, Kuperstein G, Abrahamson JL, et al: Prevalence and penetrance of germline BRCA1 and BRCA2 mutations in a population series of 649 women with ovarian cancer. Am J Hum Genet. 68:700–710. 2001. View Article : Google Scholar : PubMed/NCBI

128 

Antoniou A, Pharoah PD, Narod S, Risch HA, Eyfjord JE, Hopper JL, Loman N, Olsson H, Johannsson O, Borg A, et al: Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case Series unselected for family history: A combined analysis of 22 studies. Am J Hum Genet. 72:1117–1130. 2003. View Article : Google Scholar : PubMed/NCBI

129 

McLaughlin JR, Risch HA, Lubinski J, et al: Reproductive risk factors for ovarian cancer in carriers of BRCA1 or BRCA2 mutations: A case-control study. Lancet Oncol. 8:26–34. 2007. View Article : Google Scholar : PubMed/NCBI

130 

Liu JF, Konstantinopoulos PA and Matulonis UA: PARP inhibitors in ovarian cancer: Current status and future promise. Gynecol Oncol. 133:362–369. 2014. View Article : Google Scholar : PubMed/NCBI

131 

Ledermann J, Harter P, Gourley C, et al: Olaparib maintenance therapy in platinum-sensitive relapsed ovarian cancer. N Engl J Med. 366:1382–1392. 2012. View Article : Google Scholar : PubMed/NCBI

132 

Fujita T, Miyamoto S, Onoyama I, et al: Expression of lysophosphatidic acid receptors and vascular endothelial growth factor mediating lysophosphatidic acid in the development of human ovarian cancer. Cancer Lett. 192:161–169. 2003. View Article : Google Scholar : PubMed/NCBI

133 

Yotsumoto F, Yagi H, Suzuki SO, et al: Validation of HB-EGF and amphiregulin as targets for human cancer therapy. Biochem Biophys Res Commun. 365:555–561. 2008. View Article : Google Scholar : PubMed/NCBI

134 

Yagi H, Yotsumoto F, Sonoda K, et al: Synergistic anti-tumor effect of paclitaxel with CRM197, an inhibitor of HB-EGF, in ovarian cancer. Int J Cancer. 124:1429–1439. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 11 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sonoda K: Molecular biology of gynecological cancer (Review). Oncol Lett 11: 16-22, 2016
APA
Sonoda, K. (2016). Molecular biology of gynecological cancer (Review). Oncology Letters, 11, 16-22. https://doi.org/10.3892/ol.2015.3862
MLA
Sonoda, K."Molecular biology of gynecological cancer (Review)". Oncology Letters 11.1 (2016): 16-22.
Chicago
Sonoda, K."Molecular biology of gynecological cancer (Review)". Oncology Letters 11, no. 1 (2016): 16-22. https://doi.org/10.3892/ol.2015.3862