Epithelial ovarian tumors pathophysiology

Jump to navigation Jump to search

Epithelial ovarian tumors Microchapters

Home

Patient Information

Overview

Historical Perspective

Classification

Pathophysiology

Causes

Differentiating Epithelial Ovarian Tumors from other Diseases

Epidemiology and Demographics

Risk Factors

Screening

Natural History, Complications and Prognosis

Diagnosis

Diagnostic Study of Choice

History and Symptoms

Physical Examination

Laboratory Findings

Electrocardiogram

X-ray

Echocardiography and Ultrasound

CT scan

MRI

Other Imaging Findings

Other Diagnostic Studies

Treatment

Medical Therapy

Surgery

Primary Prevention

Secondary Prevention

Cost-Effectiveness of Therapy

Future or Investigational Therapies

Case Studies

Case #1

Epithelial ovarian tumors pathophysiology On the Web

Most recent articles

Most cited articles

Review articles

CME Programs

Powerpoint slides

Images

American Roentgen Ray Society Images of Epithelial ovarian tumors pathophysiology

All Images
X-rays
Echo & Ultrasound
CT Images
MRI

Ongoing Trials at Clinical Trials.gov

US National Guidelines Clearinghouse

NICE Guidance

FDA on Epithelial ovarian tumors pathophysiology

CDC on Epithelial ovarian tumors pathophysiology

Epithelial ovarian tumors pathophysiology in the news

Blogs on Epithelial ovarian tumors pathophysiology

Directions to Hospitals Treating Psoriasis

Risk calculators and risk factors for Epithelial ovarian tumors pathophysiology

Editor-In-Chief: C. Michael Gibson, M.S., M.D. [1]Associate Editor(s)-in-Chief: Hannan Javed, M.D.[2]

Overview

Surface epithelium of ovaries (OSE), once mistakenly referred as germinal epithelium, consists of single layer of flat to cuboidal epithelial cells. It is characterized by keratin types found in simple epithelium and functions in exchange between peritoneal cavity and the ovaries in addition to ovarian cycle. During embryonic development, surface epithelium of ovaries is a part of celomic epithelium. The future surface epithelium of ovaries then forms part of gonadal blastema and then undergoes a transformation cycle, multilayered papillary epithelium develops from simple flat to cuboidal epithelium but reverts back to simple flat to cuboidal epithelium by term. The most important functions of human surface epithelium of ovaries are its role in transport and exchange between peritoneal cavity and ovaries, and its function in repair and rupture during ovulation. Ovarian surface epithelium undergo epithelio-mesenchymal transformation to replace ovarian stroma in ovulatory repair. The previous proposition regarding the origin of epithelial ovarian cancers was that these tumors originated from surface epithelium of the ovaries and the neoplastic and metaplastic changes led to their differentiation into various histological subtypes such as serous tumors, clear cell carcinoma and endometrioid tumors. But apparent consistencies in this theory has led to development of alternate theories such as origin of neoplastic cells from fallopian tubes and endometrium.

Pathophysiology

Embryogenesis

Celomic epithelium → Peritoneal mesothelium surrounding the ovary → Metaplasia to ovarian surface epithelium[1][2]

  • During embryonic development, surface epithelium of ovaries is a part of celomic epithelium.[2]
  • Celomic epithelium itself is derived from mesothelium and forms lining of intraembryonic celom.[2]
  • The future surface epithelium of ovaries then forms part of gonadal blastema and then undergoes a transformation cycle, multilayered papillary epithelium develops from simple flat to cuboidal epithelium but reverts back to simple flat to cuboidal epithelium by term.[2]
  • It is important to note that ovarian surface epithelium is the part of celomic epithelium that overlies the presumptive gonads and the celomic epithelium in proximity of gonads also gives rise to Mullerian (paramesonephric) ducts, that in future will develop into epthelium of most of the female reproductive tract including oviducts, endometrium and a part of cervix.[2][3]
  • Ovarian surface epithelium has also been postulated to give rise or form a part of ovarian granulosa cells during embryonic development.[2][3]

Structural Characteristics of Ovarian Surface Epithelium in Adults

Cell type Surface expression Intercellular connection Basement membrane
  • Single layer
  • squamous-to-cuboidal epithelium
  • Keratin
  • Mucin antigen MUC1
  • 17β-hydroxysteroid dehydrogenase
  • Cilia
  • Simple desmosomes
  • Incomplete tight junctions
  • Integrins
  • Cadherins
  • Loosely attached
  • Tunica albuginea that is less conspicuous of its testicular counterpart
  • Keratin types that are expressed by ovarian surface epithelium are characteristic of simple epithelia such as keratin type 7, 8, 18 and 19.[2][4]
  • Catherins expressed by surface epithelium of ovaries may indicate potential for neoplastic transformation as summarized:[2][4][5][6][7]
    • Surface epithelium of ovaries typically express N-cadherin.
    • E-cadherin is typically expressed in regions where cells are columnar.
    • This selective expression of E-cadherin in regions of metaplastic epithelium may indicate propensity for neoplastic transformation.
    • P-catherin, normally absent in adult surface epithelium of ovaries, is expressed in adenocarcinoma of ovaries.

Functions

  • Two most important functions of human surface epithelium of ovaries are its role in transport and exchange between peritoneal cavity and ovaries, and its function in repair and rupture during ovulation.[2][5][8]
  • At present, its role in ovulatory rupture is not well-understood and is controversial. It is hypothesized that it contributes to follicular rupture through production of proteolytic enzymes.[2][5]
  • Epithelial need and ability of proliferation for repair of rupture and ovulatory defects is well-established and is thought to contribute the most in carcinogenesis of ovarian epithelium tumors.[2]
  • Ovarian surface epithelium undergo epithelio-mesenchymal transformation to replace ovarian stroma in ovulatory repair.[2]
  • The differentiation of surface epithelium of ovaries is, however, different from other epithelia because of its ability of differentiate into ectopic epithelium such as that of epithelium formed by Mullerian ducts.[2]

Role of Hormones and Growth Factors on Surface Epithelium

Gonadotropin-releasing hormone

and gonadotropins[2] [9]

  • Cell proliferation
Epidermal growth factor (EGF)[2] [4][10][11]
  • Cell proliferation and differentiation
  • Increased survival
Steroids[2] [12][13]
  • Decreased expression of GnRH receptors (estrogen)
  • Regulation of hepatocyte growth factor and epidermal growth factor 9estrogen)
  • Decreased expression of Transfroming growth factor β receptors (5α-dihydrotestosterone)
  • May have direct effect on proliferation stimulation
Fibroblast growth factor (FGF)[2] [14]
  • Cell proliferation
  • Increased survival
Platelet-derived growth factor (PDGF)[2] [15]
  • Cell proliferation
Tissue necrosis factor-α (TNF-α)[2] [16][17]
  • Cell proliferation
  • Increased TNFα expression
Transfroming growth factor β (TGF-β)[2] [18]
  • Decreased growth
Hepatocyte growth factor (HGF)[2] [19][20]
  • Decreased cellular adhesion
  • Increased survival and growth
Cytokines[2] [21][22]
  • Regulation of immune response
  • May increase vasculogenesis and survival

The Origin of Neoplasia in Epithelial Ovarian Cancer: A Mystery to Solve

  • The previous proposition regarding the origin of epithelial ovarian cancers was that these tumors originated from surface epithelium of the ovaries and the neoplastic and metaplastic changes led to their differentiation into various histological subtypes such as serous tumors, clear cell carcinoma and endometrioid tumors.[23][24][25][26][27]
  • However the surface epithelium of ovaries is derived from mesothelium and ovarian carcinoma resembled more closely to tissues derived from Mullerian ducts rather than ovarian mesothelium derived surface epithelium. For example serous cancer histology resembles fallopian tube epithelium and that of transitional cells tumor resembles urinary bladder. Likewise endometrioid, and sero-mucinous carcinomas are thought to have their origin in endometriosis, and Walthard nests potentially give rise to mucinous and Brenner malignant tumors, at least partially. All of these precursors are Müllerian system derivatives.[23][24][25][26][27][28]
  • Secondly the genetic profile also overcasts shadows of doubt about origin of these neoplasms from ovarian surface epithelium. The presence of identical TP53 mutations in serous tubal intra-epithelial tumors and ovarian serous tumors puts a question mark on ovarian origin theory. Gene expression profiling also demonstrated the presence of similarities between serous tubal intra-epithelial tumors and ovarian serous tumors. The various theories of origin of epithelial ovarian cancers have been discussed below.[25][26][27][29][30]

Ovarian Origin of Ovarian Epithelial Tumors

  • This simple theory states that ovarian epithelial tumors simply originate from surface epithelium of ovaries through various neoplastic changes. But recent data has highlighted the numerous inconsistencies in the theory that was once highly regarded as accurate.[25][26][27]
  • Firstly surface epithelium of ovaries is derived from mesothelium and ovarian carcinoma resembled more closely to tissues derived from Mullerian ducts rather than ovarian mesothelium derived surface epithelium. For example serous cancer histology resembles fallopian tube epithelium and that of transitional cells tumor resembles urinary bladder. Likewise endometrioid, and sero-mucinous carcinomas are thought to have their origin in endometriosis, and Walthard nests potentially give rise to mucinous and Brenner malignant tumors, at least partially. All of these precursors are Müllerian system derivatives.[23][24][25][26][27][28]
  • Secondly the presence of identical TP53 mutations in serous tubal intra-epithelial tumors and ovarian serous tumors puts a question mark on ovarian origin theory. Gene expression profiling also demonstrated the presence of similarities between serous tubal intra-epithelial tumors and ovarian serous tumors.[25][26][27][29][30]
  • The expression of PAX8 and absence of calretinin in high grade serous tumors presents another problem with theory of ovarian origin because PAX8 is a Müllerian marker and calretinin is a mesothelium marker.[26][27]
  • In 2001, a Dutch study revealed the presence of high grade serous carcinomas in fallopian tubes of women with genetic susceptibility to hereditary ovarian cancers with no evidence of such lesions in ovaries of same women. These lesions were termed as serous tubal intra-epithelial tumors.[25][26][27][31][29][32]
  • Additional studies demonstrated the presence of similar lesions in fallopian tubes of women without genetic susceptibility to ovarian cancer. In cases when fallopian tubes were removed carefully along with ovarian and/or peritoneal serous cancer, the involvement of mucosa of the tubes were found to be involved in about 70% of the cases.[25][26][27][31][29][32]
  • In an attempt to explain these apparent discrepancies it was postulated that invagination of ovarian epithelium into ovarian stroma creates “cortical inclusion cysts”. These cysts then undergo various metaplastic changes (coelomic metaplasia hypothesis) due to hormonal influence and repair mechanisms to give rise to ovarian epithelial cancer. Although these cysts are present but no such neoplastic and metaplastic transformation has been reported or observed until now. Additionally the observed cysts could dimply be the transplants from the fallopian tubes.[25][26][27][33]
  • Another proposed theory is the implantation of tubal epithelium from fimbria into ovarian inclusion cysts due to their close contact during the ovulation process. This may explain the origin of serous tumor of the ovaries but unable to explain other tumor sub-types.[25][26][27]

Secondary Müllerian System

  • This theory tries to explain this apparent enigma of existence of Müllerian epithelial lesions in locations not derived from Müllerian ducts such as ovaries and peritoneal cavity. Secondary Müllerian system consists of müllerian-type tissue lined cysts that are located in close proximity to the ovaries.[34][25][26][27]
  • According to this hypothesis Müllerian tissues, considered by some as vestigial, are found in locations such as para-tubal and para-ovarian locations and these tissues or cysts, not the ovarian epithelium itself, give rise to epithelial ovarian neoplasms. These tumors, arising outside ovaries, then enlarge and become implants/or compress ovaries and present as ovarian tumors.[23][25][26][27]
  • But there are number of problems this theory fails to explain. For example mucinous epithelial tumors of ovaries resemble intestinal epithelium rather than endocervical epithelium. Also transitional cell tumor resemble morphologically to bladder epithelium that is not a derivative of Müllerian system.[25][26][27][28]
  • Another apparent flaw is that transition of these cysts lined by Müllerian-type epithelium, although present, to carcinoma has been very rare.[25][26][27]

The Origin of Epithelial Ovarian Tumors from Fallopian Tubes and Endometrium

  • The evidence from recent studies indicate that majority of epithelial ovarian cancers have their origin outside ovaries, especially from fallopian tubes and endometrium. This idea is supported by several observations in a number of studies.[25][26][27]
  • The histology of serous, endometrioid and clear cell carcinoma demonstrates that their morphology is similar to that fallopian tubes, and endometrium rather than ovarian epithelium.[25][26][27][35][36]
  • Presence of PAX8, a Müllerian marker, and absence of calretinin, a mestothelial marker, further supports the theory. Moreover the genetic profile expression similarities and presence of similar TP53 mutation signatures in serous tubal intra-epithelial tumors and epithelial ovarian cancers also supports the extra-ovarian origin of epithelial ovarian cancer.[25][26][27][29][30].
  • In 2001, a Dutch study revealed the presence of high grade serous carcinomas in fallopian tubes of women with genetic susceptibility to hereditary ovarian cancers with no evidence of such lesions in ovaries of same women. These lesions were termed as serous tubal intra-epithelial tumors.[25][26][27][29][32][31]
  • Additional studies demonstrated the presence of similar lesions in fallopian tubes of women without genetic susceptibility to ovarian cancer. In cases when fallopian tubes were removed carefully along with ovarian and/or peritoneal serous cancer, the involvement of mucosa of the tubes were found to be involved in about 70% of the cases.[25][26][27][29][32][31]
  • These tubal serous lesions were located in fimbria in almost all of the cases, giving rise to the proposition that serous tumors originated in fallopian tubes and then implantation into ovaries.[25][26][27][37][29][38]
  • The association between adnexal malignant mixed mesodermal tumors and serous tubal intraepithelial tumors pints further in direction of tubal origin of these epithelial ovarian tumors.[25][26][27][39][40]
  • Similarly morphologic and molecular studies have indicated that endometrioid and clear cell carcinoma of the ovaries have their origin in endometriosis. These studies suggest that these tumors arise from endometriomas, the endometriotic cysts that are present outside the normal endometrium.[25][26][27][35][36]
  • This theory regarding the origin of endometrioid and clear cell carcinoma of the ovary is supported by the fact that tubal ligation that prevents endometriotic implants into ovary and peritoneum in endometriosis has a protective effect on endometrioid and clear cell type cancers but not on the serous cancer of the ovary because it doesn't occlude the connection between fimbria and the ovaries.[25][26][27][41][42]

The Origin of Mucinous Tumors of Gastrointestinal Type and Transitional Cell (Brenner) Tumors: Still a Mystery to Solve

  • Mucinous and the transitional tumors of ovaries are two of the least common types of the epithelial ovarian tumors. In fact, most of the mucinous tumors in ovaries are secondary and primary tumors only form about 3% of all epithelial ovarian cancers. Mucinous epithelium in mucinous tumors of ovaries resemble more to intestinal mucinous epithelium rather than that of endocervix as was previously argued. Transitional cell tumors, on the other hand, are closer to bladder epithelium in histological studies.[27][25][26][28][43][44]
  • Another study demonstrated the presence of Brenner tumor foci in mucinous cystadenoma in almost one fifth of the cases. Alternatively the association of mucinous tumors with Walthard cell nests, which are composed of transitional-type epithelium, also indicates the connection between mucinous and transitional tumors.[27][25][26][43][44]

Pathogenesis

DNA damage and repair mechanisms.
DNA damage and repair mechanisms.[45]

Secondary Müllerian System

  • Although ovarian surface epithelium is not a derivative of Müllerian ducts but ovarian epithelial cancers are characterized by presence of Müllerian lesions.[34]
  • Serous carcinoma of ovary is though to originate from fallopian tubes while clear cell, endometrioid, and sero-mucinous carcinomas are thought to have their origin in endometriosis. Similarly Walthard nests potentially give rise to mucinous and Brenner malignant tumors, at least partially. All of these precursors are Müllerian system derivatives..[23][24]
  • Secondary Müllerian system is a hypothesis that tries to explain this apparent enigma of existence of Müllerian epithelial lesions in locations not derived from Müllerian ducts such as ovaries and peritoneal cavity.[46][24]
  • According to this hypothesis, Müllerian tissues, considered as vestigial, are found in locations such as para-tubal and para-ovarian locations and these tissues or cysts, not the ovarian epithelium itself, give rise to epithelial ovarian neoplasms.[23][24]

Hereditary Ovarian Carcinoma: An Understanding of Genome

  • More than one fifth cases of ovarian epithelial cancers are found to have hereditary causes. These hereditary diseases/syndromes appear to possess heterogeneous, both in genetic anomalies and in clinical manifestations.[47][48]
  • Majority of these hereditary cancers are caused by two genetic anomalies: a defect in so-called mismatch repair genes named as MLH1, MSH2, MSH6 and PMS2, and in DNA defects repair genes named as BRCA1 and BRCA2.[47][48][45][49]

The Role of BRCA1 Gene in DNA Repair

  • BRCA1 is a protein that, through a complex interaction with other proteins such as tumor suppressors, regulators of cell cycle and other DNA repair genes, is involved in DNA repair pathways.[47][48] [50]
  • This protein has two domains: amino-terminal RING domain and a BRCT domain. The former posses E3 ubiquitin ligase activity and the later facilitates phospho-protein binding.[50]
  • Tumor suppressor role of both domains is highlighted by the fact that mutations in both domains have been found in breast and gynecological malignancies.[50]
  • The major role of BRCA1 appears to sense and repair double stranded DNA breaks in homologous recombination.[50]

Binding of BRCA1 to double stranded DNA breaks through its association with the abraxas–RAP80 macro-complex → processing of double stranded DNA breaks through interaction of BRCA1 with CtIP (transcription factor) and the MRN complex → The BRCA1–CtIP complex → CtIP-mediated 5′-end resection of double stranded DNA breaks

  • Another role of BRCA1 in Non-homologous end joining (NHEJ) pathway has also been proposed. Though still controversial, it has been suggested that BRCA1 plays a critical function by removal of Non-homologous end joining proteins such as p53-binding protein 1 (53BP1) from double stranded DNA breaks.[50]
  • G1/S, S-phase and G2/M checkpoints activation during cell cycle has also been found defective in cells lacking or having mutated BRCA1. A brief interaction of BRCA1 with cell cycle is given below:[50]

Phosphorylation of BRCA1 by ataxia telangiectasia mutated (ATM) or ataxia telangiectasia and Rad3-related protein (ATR) → phosphorylation of p53 → transcriptional induction of the cyclin dependent kinase (CDK) inhibitor p21.

[51]A summary of BRCA1 activity and function in DNA damage repair

The Role of BRCA2 Gene in DNA Repair

  • BRCA2, as opposed to BRCA1 that functions in multiple pathways involving DNA repair, has its primary role in homologous recombination (HR).[47][48][50]
  • DNA-binding domain (DBD) of BRCA2 binds single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA) and eight BRC repeats. The eight BRC repeats bind RAD51 (a recombinase).[50][52]
  • The binding of BRCA2 to RAD51 leads to recruitment of RAD51 to double stranded DNA breaks, an essential step in homologous recombination double stranded DNA repair.[50][52][53]
  • After recruitment, BRCA2 helps RAD51 in displacement of replication protein A (RPA) in single stranded DNA. It then prevents nucleation of RAD51 at double stranded DNA and promotes RAD51 filament formation on single stranded DNA.[50][53]

The Connection Between BRCA1 and BRCA2

  • The common pathway that seems to link both BRCA! and BRCA2 proteins is homologous recombination mediated repair.[50][54][55]
  • Partner and localizer of BRCA2 (PALB2) physically connects BRCA1 and BRCA2 through N-terminal coiled-coil domain and the C terminus.[50][54][55]
  • The interaction between BRCA2 and PALB2 is observed for two critical function in homologous recombination mediated repair: interaction of RAD51 with replication protein A (RPA) in single stranded DNA and recruitment of BRCA2 and RAD51 on the site of DNA damage.[50]

The Role of Mismatch Repair Genes

  • Mismatch repair genes mutated in pathogenesis of hereditary epithelial ovarian cancer include human MutS homolog (MSH2 and 6), the human MutL homolog (MLH1 and 3), and post-meiotic segregation MutL homolog (PMS2) genes.[47][48][45][49]
  • A simplified version of repair mechanism by mismatch repair genes products is described below:[49][56]

MutS homologs (MSHs) recognize the DNA mismatch → MutS homologs (MSHs) recruit MutL homologs (MLHs) → excision of mismatched DNA → DNA polymerase re-synthesizes DNA.

  • Cells deficient in mismatch repair mechanism develop high rate of mutations including DNA sequences that include microsatellite repeats, resulting in microsatellite instability. This microsatellite instability has been implicated in impaired or defective signaling transduction, DNA repair and apoptosis, transcriptional regulation and protein translocation, and immune regulation.[45][49][56]
Mismatch repair genes and oncogenesis.
Mismatch repair genes and oncogenesis. Source: Courtesy of G. Multoff and J. Radons, Frontiers in oncology

TP53 Mutations and Loss of Tumor Suppression

  • TP53 is a tumor suppressor gene that encodes for a transcription factor. The transcription factor encoded by TP53, known as p53, is a major regulator of cell cycle.[57][58]
  • Called by some as “Guardian of the Genome”, it is involved in variety of cellular functions such as cellular proliferation and cell cycle, apoptosis, and stability & integrity of the genome.[59][58]
Downstream effect of p53 mutation
Downstream effect of p53 mutation.[60]

Template:Epithelial ovarian cancer

An Insight Into The Molecular Pathogenesis of Epithelial Ovarian Cancer

Genetic alterations in cell cycle genes in epithelial ovarian cancer types.
Genetic alterations in cell cycle genes in epithelial ovarian cancer types.[61]

Dualistic Model

  • This model attempts to explain clinicopathological and molecular genetic features of epithelial tumors by diving them in two subgroups: type I and type II epithelial ovarian tumors.[25][62]
  • Another advantage of this classification is that it tries to group precursor lesions with their putative malignant lesions.[25][62]
  • Type I tumors generally arise from endometriosis or fallopian tubal related serous epithelium. They are clinically stable, exhibit less aggressive clinical course and a different genetic than that of Type II.[63][64]
  • Type II tumors generally arise from fallopian tubal epithelium. They exhibit more aggressive clinical course and a different genetic profile relative to Type I.[63][64]
  • Type I tumors are generally characterized by chromosomal stability and somatic mutations that may include KRAS, BRAF, PTEN, PIK3CA, CTNNB1, ARID1A and PPP2R1A. BRCA1 mutation, on the other hand, has not been observed and TP53 mutation is very rare.[25][62][65]
  • Type II tumors are characterized by chromosomal instability. The mutations characteristic of high grade tumors, especially TP53 are common. TP53 has been reported in more than 90% of these tumors and a high proportion contains either BRCA mutations or BRCA related mutations such as RAD51, PALB2.[25][66][67]
  • A simplified version of this classification is provided below:
Epithelial Ovarian Cancer
Type I Type II
  • Low-grade serous carcinoma
  • Endometrioid carcinoma
  • Clear cell carcinoma
  • Mucinous carcinoma
  • Malignant Brenner tumor
  • Seromucinous carcinoma
  • High-grade serous carcinoma
  • Undifferentiated carcinoma
  • Carcinosarcoma

Dualistic Model for Serous Tumor

  • Serous tumor provides, perhaps the most, evidence for the proposed model. Studies suggest that it exhibits distinct morphological and genetic types/stages that may explain the progression from benign tumor (cystadenoma) to low grade serous tumor.[25][62]
  • This idea is supported by advances in discovery and understanding of so-called borderline serous tumors. These advances demonstrated that one type of these borderline tumors resembled benign serous tumors in their cinicopathological behavior and were named as “atypical proliferative serous tumor (APST)”. The other type behaved in way closer to low grade serous cancer and were termed as “micropapillary serous carcinoma (MPSC)”.[25][62][68]
  • The absence of KRAS and BRAF mutation in serous cystadenoma but presence of these mutations in atypical proliferative serous tumor indicates that these mutations occur somewhat early in transformation of serous cystadenoma into atypical proliferative serous tumor.[25][69]
  • More support was provided by studies that showed that genes involved in MAPK pathway were expressed more in micropapillary serous carcinoma than in atypical proliferative serous tumor. In addition, micropapillary serous carcinoma exhibited more chromosomal instability than atypical proliferative serous tumor.[25][68]
  • This indicates the step-wise development of low grade serous carcinoma from benign cystadenoma with developemnet of abnormalities in KRAS, BRAF and MAPK pathways. A simplistic version is given below:[25][70][71][72]

ERRB2 (mutation) → PI3K → AKT → mTOR → Cyclin D1 → cell cycle control and cellular survival → Tumor initiation and progression

KRAS → BRAF → MEK → ERK → Cell cycle control and cellular survival → Tumor initiation and progression

PI3K (mutation) → AKT → Tumor initiation and progression

KRAS (mutation) → BRAF → MEK → ERK → Tumor initiation and progression

PI3K (mutation) → Tumor initiation and progression

BRAF (mutation) → MEK → ERK → Cell cycle control and cellular survival → Tumor initiation and progression

*ERK can directly promote tumor initiation, and cellular growth and survival or can promote these through activation of glucose transporter-1 and cyclin D1.[25]

  • High grade serous carcinoma, on the other hand, is characterized by mutations rarely found in either of low grade serous carcinoma, micropapillary serous carcinoma and atypical prolferative serous tumor. Of these mutations, TP53 is the most common mutation and is found in >90% of the cases.[25][67]
  • While BRCA1 and BRCA2 mutations occur in majority of familial high grade serous carcinoma, inactivation of BRCA1 and/or BRCA2 by indirect mechanisms such as mutation and/or inactivation of promoter occur more frequently in sporadic high grade serous cancer and have been observed in about half of these cancers.[25][73]
  • The most noteworthy feature in molecular pathogenesis of high grade serous carcinoma is high level of DNA copy number gains or losses. These gains or losses are diffuse and include foci such as CCNE1 (cyclin E1), NOTCH3, AKT2, RSF1, and PIK3CA.[25][74]
Pathogenesis of high grade serous carcinoma
Pathogenesis of high grade serous carcinoma[75] Normal fallopian tube epithelium comprises of both secretory and ciliated cells and stains negative for p53. The benign ‘p53 signature’: secretory cells that possess strong p53 expression and evidence of DNA damage but are not proliferative. When they progress to serous tubal intraepithelial carcinoma or ‘STIC’, they acquire nuclear pleomorphism, mitoses, and loss of polarity. Serous tubal intraepithelial carcinoma shares all these properties with invasive high grade serous epithelial ovarian cancer and clinical symptoms typically emerge with advanced disease.[75][76]

Genetic Alterations in Clear Cell

  • Inactivating mutation of ARID1A. ARID1A encodes for a product that functions in tumor suppression and is observed in half of clear cell cancers.[25][65][77]
  • Activating mutation of PIK3CA, also observed in about half of these tumors, results in actiavtion of PI3k pathway.[25][78]
  • Deletion of PTEN, observed in about 20% of the cases, results in loss of tumor suppressor gene.[25][79]
  • These alterations indicate the importance of PI3K/PTEN pathway in development of clear cell carcinoma of ovary.[25][79]
ARIDA loss and PIK3CA activation in clear cell cancer of ovaries.[80](A) ARID1A and PIK3CA alterations plot against TCGA datasets. Significance of association between ARID1A and PIK3CA mutations were determined using Fisher’s exact test. (B) Determination of CRE-deleted (Arid1aΔ) allele in samples of tumor DNA. (C) RT-PCR was used to detect ARID1A loss or (Gt)Rosa26Pik3ca*H1047R transcripts. (D and E) Expression of ARID1A in normal ovaries (E) Expression of ARID1A in the normal ovarian surface epithelium (arrowhead). (F) ARID1A expression is not observed in the tumors. (H, I) Highest expression of P-AKT S473 in surface epithelium of ovaries in normal ovaries (E, arrowhead) and are greatly increased in ovarian tumors (F, arrowhead). Asterisk in E denotes an oocyte. (J,K) Morbid Arid1afl/fl;(Gt)Rosa26Pik3ca*H1047R mouse at sacrifice with hemorrhagic ascites (inset), primary ovarian tumor of moderate size, and bilateral tumor metastases (arrowheads). (L,M) Morbid Arid1afl/fl;(Gt)Rosa26Pik3ca*H1047R mouse at sacrifice with hemorrhagic ascites (inset), large primary ovarian tumor, and no visible metastases. The mice shown in J-M were sacrificed at 7 and 9 weeks post-AdCRE, respectively, because of visible ascitic fluid burden. (N,O) Arid1afl/+;(Gt)Rosa26Pik3ca*H1047R mice at 11-weeks post-AdCRE showing no evidence for tumor formation. In K and M, dashed circles indicate primary ovarian tumor on injected ovary. In N, arrows denote the AdCRE injected ovary. In K, M, and O, asterisks denote the uninjected, control ovary.

Genetic Alterations in Endometrioid Tumors

  • Low grade endometrioid cancer also exhibits dysregulated either PI3K/PTEN pathway or Wnt/b-catenin signaling pathway. Later has been observed in about 40% of the low grade endometrioid tumors.[25][81][82][83]
  • PI3K/PTEN pathway is deregulated either by activating mutations in PIK3CA or inactivation/deletion of PTEN, a tumor suppressor gene. Activating mutations of CTNNB1, that encodes β-catenin, are usually the cause for deregulated Wnt/b-catenin signaling pathway.[25][81][82][83]
  • High grade endometrioid carcinoma, on the other hand, dooes not exhibit dysregulated PI3K/PTEN pathway or Wnt/b-catenin signaling pathway but frequently has TP53 mutations present.[25][83]

Genetic Alterations in Mucinous Tumors

  • KRAS mutations are present in up to two thirds of these tumors and have also been used as molecular marker.[25][84][85]
Possible genetic alteration in epithelial ovarian cancers
Protein Normal function Function in malignancy
Human Epidermal growth factor receptor (HER-1)[86][87]
  • Promotes cell proliferation
  • Opposes apoptosis
  • Regulates differentiation
  • Activating mutation
  • Increased cellular proliferation
  • Inhibition of apoptosis
Human Epidermal Growth Factor Receptor 2 (HER-2)[86][87]
  • Promotes cell prolifeartion
  • Inhibition of apoptosis
  • Regulates differentiation
  • Activating mutation
  • Increased cellular proliferation
  • Inhibition of apoptosis
Non-receptor tyrosine kinase Src[88][89] Involved in regulation of
  • Gene transcription
  • Angiogenesis
  • Cellular adhesion
  • Cellular proliferation
  • Activating mutation
  • Increased angiogenesis
  • Decreased cellular adhesion
  • Increased tumor metastasis
  • Increased cellular proliferation
Colony stimulating factor-1/fms[1][90][91]
  • Increased macrophage survival
  • Increased macrophage proliferation
  • Increased macrophage differentiation
  • Activating mutation
  • Stimulation of tumor cell proliferation
  • Increased angiogenesis
  • Promotes tumor invasion
  • Increased metastasis
  • Decreased anoikis
Insulin-like growth factor/receptor ILGF/ILGFR[92][93][94]
  • Promotes growth
  • Increased survival
  • Activating mutation
  • Increased proliferation
  • Enhanced survival
  • Suppression of cell cycle regulators
k-ras[95][96]
  • Cellular proliferation
  • Cell survival
  • Activating mutation
  • Increased proliferation
  • Enhanced survival
b-raf[97][98]
  • Cellular proliferation
  • Cellular differentiation
  • Activating mutation
  • Increased proliferation
  • Enhanced growth
Transforming growth factor-β[99][100][101]
  • May function as a tumor suppressor and a promoter
  • Promotes growth arrest
  • Maintains cellular homeostasis
  • Increased proliferation
  • Decreased apoptosis
  • Epithelial-to-mesenchymal transition
  • Sustained angiogenesis
  • Evasion of immune surveillance
  • Metastasis
myc[102][103][104]
  • Derives cellular proliferation
  • Increased growth
  • Cell-cycle mediator
  • Inhibits apoptosis
  • Stem-cell renewal
  • Activating mutation
  • Increased proliferation
  • Decreased apoptosis
  • Increased metabolism in tumor cells
Cyclin D/Cdk4/6[105][106][107]
  • Cell-cycle mediator
  • Controls G1 length
  • Activating mutation
  • Decreased G1 length
  • Increased proliferation
  • Increased angiogenesis
Cyclin E/Cdk2[108][109][110]
  • Cellular proliferation
  • Cell-cycle mediator
  • Assembly of the pre-replication complex
  • Promotes G0 to cell cycle entry
  • Promotes G1 to S phase transition
  • Decreased apoptosis
  • Activating mutation
  • Increased cellular proliferation
  • Impaired apoptosis
  • Increased cellular survival
Cyclin B/Cdk1[111][112][113]
  • Cell-cycle mediator
  • promotes G2 to M phase transition
  • Activating mutation
  • Increased cellular proliferation
  • Promotes malignant transformation
p16[114][115][116]
  • Member of the INK4 family of CDK inhibitors
  • Inhibits Cyclin D/Cdk4/6
  • Decreased G1 to S phase transition
  • Lost or downregulated
  • Decreased G1 length
  • Increased proliferation
  • Increased angiogenesis
p27 (kip-1)[117][118][119]
  • Inhibitor of Cyclin E/Cdk2
  • Mediates cell cycle arrest
  • Decreased G1 to S phase transition
  • May act as oncogen and promote proliferation
  • Lost or dysregulated
  • Increase in cell proliferation
  • Impaired apoptosis
p21 (WAF-1)[120][121][122]
  • Inhibits cyclin-dependant kinases
  • Cell-cycle arrest
  • Decreased proliferation
  • Promotes cellular differentiation
  • May inhibit/promote apoptosis
  • May act as oncogen and promote proliferation
  • Lost or dysregulated
  • Increase in cell proliferation
  • Decreased cellular differentiation
  • Decreased apoptosis
  • Correlates positively
    • tumour grade
    • invasiveness
    • aggressiveness
Nuclear factor-κB[123][124][125]
  • A transcription factor involved in regulation of
    • immune response to inflammation
    • expression of cytokines, chemokines, and adhesion molecules
    • cell cycle
    • apoptosis
  • May function as a tumor suppressor and a promoter
  • Dysregulated
  • Increased angiogenesis
  • Enhanced tumor growth
  • Induces resistance to chemotherapy by acting as anti-apoptosis
NOEY(ARHI)[126][127][128][129]
  • Inhibits cell growth
  • Induces apoptosis
  • Inhibits tumor cells migration through chemotaxis and haptotaxis
  • Inactivating mutation
  • Enhanced tumor growth
  • Decreased apoptosis
  • Increased chances for metastasis
PIP3/Akt[130][131]
  • Akt is activated by PIP3 and plays a role in
    • regulation of cellular growth
    • cell cycle progression
    • regulation of glucose metabolism
    • genome stability
    • gene transcription
    • protein synthesis
    • neovascularization
    • promotes cell survival by blocking apoptosis
  • Activating mutations
  • Increased cellular proliferation
  • Increased tumor cells survival
  • Increased tumor cells migration
  • Increased tumor cells invasion
  • Chemotherapy resistance
  • Decreased apoptosis
  • May promote angiogenesis
PTEN[132][133][134]
  • Suppresses Akt and thus regulates cell cycle, cellular growth and apoptosis
  • Regulates self-renewal and differentiation of human stem cells
  • Regulates oocyte growth and follicular activation
  • Regulates chemotaxis of neutrophils
  • Inhibit cell invasion and migration
  • Deletion or inactivating mutation
  • Increased cellular proliferation
  • Increased tumor cells survival
  • Increased tumor cells migration
  • Increased tumor cells invasion
  • Decreased apoptosis
p53[135][136][137]
  • A transcription factor that
    • regulates cell cycle
    • promotes DNA damage repair
    • promotes apoptosis
    • maintains genomic integrity
  • Loss results in
    • DNA damage and carcinogenesis
    • increased tumor cell growth and survival
    • increased metastasis
    • decreased apoptosis
    • resistance to chemotherapy
BRCA1[50][48][138]
  • A tumor suppressor that mediates double stranded DNA repair through
    • homologous recombination pathway
    • non-homologous end joining pathway
  • Activates checkpoints in cell cycle
  • Maintains genomic integrity
  • Mutations are responsible for hereditary breast & ovarian tumors
  • Loss results in
    • DNA damage and carcinogenesis
    • increased tumor cell growth and survival
BRCA2[50][48][138]
  • A tumor suppressor that mediates double stranded DNA repair through
    • homologous recombination pathway
  • Maintains genomic integrity
  • Protects replication fork and replication fidelity
  • Mutations are responsible for hereditary breast & ovarian tumors
  • Loss results in
    • DNA damage and carcinogenesis
    • increased tumor cell growth and survival
  • Defects in maintenance the length of the nascent strand of DNA
MLH1/MSH2[139][140][141]
  • Tumor suppressors that
  • mediates DNA damage repair
  • maintains genomic integrity
  • possible regulation of cell cycle
  • Loss results in
    • DNA damage and carcinogenesis
    • increased survival
    • resistance to chemotherapy
    • chromosomal instability
    • microsatellite instability (MSI)
    • the cytosine phosphate guanine (CpG) island methylator phenotype (CIMP)
Fas ligand[142][143][144]
  • Binds to Fas receptor and induces apoptosis
  • Expressed mainly on T-lymphocytes
  • May induce apoptosis in cancer cells and virus infected cells
  • May also be involved in
    • liver regeneration following partial hepatectomy
    • neurite outgrowth
  • Most tumor cells are resistant to Fas-FasL mediated apoptosis
  • Tumor cells express FasL to induce apoptosis in cytotoxic lymphocytes
  • Promotes tumor cells survival
  • Enhances tumor cells invasion
  • Increased tumor cells migration
Human leukocyte antigen-G[145][146][147]
  • Inhibits T-cell function through
    • inhibiting proliferation
    • causing cytotoxicity
    • inducing apoptosis
    • cytokine production in B lymphocytes
    • inhibiting differentiation
  • Inhibits proliferation and cytotoxicity of natural killer cells
  • Promotes angiogenesis
  • Inhibits chemotaxis
  • Promotes progression of cancer through evasion of immune response by
    • inhibiting T-cell functions by inducing apoptosis and decreased proliferation
    • inhibiting T-cell differentiation through various mechanisms
  • Inhibits proliferation and cytotoxicity of natural killer cells
  • Promotes angiogenesis
  • Inhibits chemotaxis of cytotoxic cells
hTERT[148][149][150]
  • Maintains telomeres length
  • Promotes replication
  • Up-regulated in majority of human cancers
  • Provides limitless replication ability to cancer cells
Vascular endothelial growth factor/Vascular endothelial

growth factor receptor[151][152][153]

  • Stimulates angiogenesis through
    • increased endothelial cell survival
    • Increased endothelial cell proliferation
    • increased endothelial cell migration
  • Increases vascular permeability
  • May regulate fibroblasts in the stroma of tumors
  • May effect tumor stem cells
  • Promotes angiogenesis
  • Promotes tumor cells growth
  • May initiate carcinogenesis
  • Promotes invasion and metastasis of tumor cells
Interleukin-8[154][155][156]
  • Chemokine produced to recruit leukocytes and myeloid-derived suppressor cells
  • Promotes epithelial-to-mesenchymal transition
  • Promotes infection resolution
  • Promotes angiogenesis
  • Promotes epithelial-to-mesenchymal transition in tumor cells
  • Promotes resistance to chemotherapy
  • Tumor progression through immunosuppressive and pro-tumorigenic immune cells
  • Promotes angiogenesis
  • Promotes invasion and metastasis
EphA2[157][158][159]
  • Promotes angiogenesis
  • Plays a key role in development of
    • Lens
    • Inner ear
    • Mammary glands
  • Promotes kidney repair following injury
  • Promotes bone remodeling bone remodeling
  • Over-expressed in ovarian epithelial cancer
  • Promotes tumor initiation
  • Promotes neo-vascularization
  • Promotes tumor invasion
  • Promotes metastasis
Matrix metalloproteinases[160][161][162]
  • Proteases that degrade tissues, matrix and other proteins and play a role in
    • bone modeling and remodeling
    • mammary development
    • blood vessels remodeling
    • a variety of other tissues such as tracheal tube
  • Promotes inflammation through enzymatic activation
  • Over-expressed in ovarian epithelial cancer
  • Promotes tumor invasion through degradation of extra-cellular matrix
  • Promotes metastasis through degradation of extra-cellular matrix
  • May have a role in tumor initiation and angiogenesis
αvβ3[163][164][165]
  • One of the most important mediator of angiogenesis
  • promotes smooth muscle cells migration and proliferation
  • Promotes angiogenesis
  • Promotes survival
Focal adhesion kinase (FAK)[166][167][168]
  • Promotes endothelial cells migration
  • May play a role in integrin-dependent cell survival signal
  • Inhibits apoptosis
  • Enhances cell motility
  • Promotes angiogenesis
  • Promotes tumor cells survival
  • Inhibits apoptosis
  • Promotes tumor metastasis
E-cadherin[169][170][171]
  • One of the most important promoter of cell-cell adhesion
  • Play critical role in formation and maintenance of epithelia, and tissue formation
  • Loss or mutations results in
    • epithelial–mesenchymal transition
    • decreased cell-cell adhesion
    • tumor cells invasion
    • metastasis

Hereditary Epithelial Ovarian Carcinoma: An overview of Hereditary Syndromes and the Genetic Mutations

Hereditary Breast and Ovarian Cancer (HBOC)

  • Hereditary breast and ovarian cancer (HBOC) is an autosomal dominant disorder caused by mutations in BRCA1 and BRCA2 genes that are responsible for DNA repair in homologous recombination pathway.[47][50]
  • Individuals with this disorder are at risk of developing breast (lifetime risk is 30-80%) and ovarian cancer (lifetime risk is 30-50%), along with other malignancies such as pancreatic, stomach, laryngeal, fallopian tube and prostate cancer.[47][50]
  • The reason for increased susceptibility to ovarian and epithelial cancer is not fully understood but but may be explained by repression of the transcription of hormone-mediated signalling factors or production of reactive oxygen species during menstrual cycle mediating DNA damage.[50][172][173]
Malignancies associated with BRCA mutations (Hereditary breast and ovarian cancer syndrome)[174]
  • Breast cancer (male and female)
  • Ovarian cancer
  • Fallopian tube carcinoma
  • Primary papillary carcinoma of the peritoneum
  • Prostate cancer
  • Uterine body cancer
  • Cervical cancer
  • Pancreatic cancer
  • Gall bladder cancer
  • Bile duct cancer
  • Stomach cancer
  • Melanoma

Lynch Syndrome

  • Lynch syndrome (LS), also known as hereditary nonpolyposis colon cancer (HNPCC), is characterized by germline mutations in DNA mismatch repair genes MLH1, MSH2, MSH6, MLH3, and PMS2.[47][45][49]
  • A simplified version of repair mechanism by mismatch repair genes products is described below:[49][56]

MutS homologs (MSHs) recognize the DNA mismatch → MutS homologs (MSHs) recruit MutL homologs (MLHs) → excision of mismatched DNA → DNA polymerase re-synthesizes DNA.

  • Accounted for 10-15% of all ovarian cancers, this syndrome is caused by inherited mutation in one allele and then loss of second allele (secondary hit).[47][175]
  • The most common malignancies in Lynch syndrome are colorectal carcinoma and gynecological cancers, endometrial carcinoma being the most common among gynecological malignancies followed by ovarian carcinoma.[175]
  • Other malignancies that have been observed in lynch syndrome are gastric cancer, small bowel malignancies, hepatobiliary epithelial carcinoma, uroepithelial epithelial carcinoma and brain tumors.[175][176]
Genetic variation in Lynch syndrome.
Genetic variation in Lynch syndrome.[177]

Li-Fraumeni Syndrome

  • Li-Fraumeni Syndrome is an autosomal dominant disorder caused by germline mutation in TP53, the most mutated gene in human cancers. The most common of the mutations are missense mutations.[57][58]
  • TP53 encodes for a transcription factor that responds to various cell signals and is a major regulator of the cell cycle. It is involved in variety of cellular functions such as cellular proliferation and cell cycle, apoptosis, and stability & integrity of the genome.[59][58]
  • Mutations in TP53 resulting defective or decreased p53 are not only implicated in pathogenesis but also impact prognosis, causing worse survival rate among the individuals with the mutations.[59][178]
  • These mutations are most commonly observed in epithelial ovarian cancer (47%), colorectal carcinoma (43%), head/neck cancer (42%), and esophageal cancer (41%). Breast cancer, sarcoma and brain, and adrenocortical carcinoma account for majority of the tumors encountered in Li-Fraumeni syndrome.[59][179]

Site-Specific Ovarian Cancer

  • A term used to describe families in which there are several relatives with epithelial ovarian cancer but no other co-existent malignancies that are associated with other hereditary syndromes associated with epithelial ovarian cancer.[180]
  • A hypothesis is that it is caused by gen/genes that are yet to be identified. Site-specific ovarian cancer appears to be transmitted in autosomal-dominant fashion in some families but some studies have suggested the risk to be as low as 5%.[180][181]

Cowden Syndrome

  • An autosomal-dominant syndrome , caused by mutations in PTEN gene, has been associated with a variety of neoplastic/non-neoplastic lesions and clinical manifestations throughout the body including:[180][182][183]
    • Epithelial ovarian cancer
    • Hamartomatous lesions of skin and organs
    • Macrocephaly
    • Breast cancer
    • Thyroid cancer
    • Endometrial cancer

RAD51

  • RAD51 is a recombinase that binds with eight BRC repeats of BRCA2. This allows RAD51 to be recruited to double stranded DNA breaks, an essential step in homologous recombination double stranded DNA repair.[50][52][53][57]
  • Some studies have suggested risk for developing ovarian cancer in RAD51 mutations is as high as six-fold. There is also an increased risk for developing breast cancer.[57][184][185]

PALB2

  • Partner and localizer of BRCA2 (PALB2) physically connects BRCA1 and BRCA2 through N-terminal coiled-coil domain and the C terminus. This BRCA2 interacting protein plays an essential role in DNA repair.[50][54][55]
  • The association of PALB2 with ovarian cancer has not be fully established but an increased risk for breast cancer, pancreatic cancer and ovarian cancer has been observed in some studies.[45][186][187]

CHEK2

  • CHEK2 gene encodes for a protein called checkpoint kinase 2 (CHK2). It interacts with other regulators and tumor suppressors such as TP53 to play a role in tumor suppression through cell-cycle regulation and apoptosis.[188][189]
  • There are conflicting results regarding association of CHEK2 with ovarian cancers. Some studies have suggested no association but the limitations were observed because of focus on only certain allelic mutations in CHEK2.[45][190]

Mre11 Complex

  • Mre11 Complex is involved in DNA repair and comprises of meiotic recombination 11 (MRE11), RAD50 and Nijmegen breakage syndrome 1 (NBS1; also known as nibrin).[45][191]
  • This complex plays an essential role in homologous recombination mediated DNA repair, non-homologous end-joining (NHEJ) and alternative non-homologous end-joining (A-NHEJ) pathways, all involved in double stranded DNA repair.[191][192]
  • Some studies have suggested an increased susceptibility to ovarian and breast cancers in hereditary mutations in Mre11 complex.[45][193]

BARD1

  • This gene encodes for a peptide that interacts with BRCA1 and forms a heterodiamer that plays a role in homologous recombination mediated repair of double stranded DNA breaks.[194][195]
  • Mutations in BARD1 have been associated with breast and ovarian cancer.[45][196]

BRIP1

  • BRCA1-interacting protein 1 (BRIP1) encodes for a helicase that interacts with BRCA1 in homologous recombination mediated repair of double stranded DNA breaks.[197][198]
  • Mutation in BRIP1 gene association with familial ovarian cancer have been demonstrated in some studies. There also been proposed risk for breast cancer but it has yet to be established.[198][199]

An attempt to explain the origin of carcinogenesis in sporadic epithelial carcinoma

Proposed

hypothesis

Proposed

Mechanism

For Against
Incessant ovulation[1][200][201][202][203][204]
  • Every ovulatory cycle leads to epithelial injury and resultant repairs make cells more susceptible to mutations
  • Increased incidences of ovarian epithelial cancers in advanced age (increased number of cycles)
  • Factors that decrease ovulatory cycles such as oral contraceptive use, pregnancy and breast-feeding decrease the risk for ovarian epithelial cancer
  • Progesterone only oral contraceptives do not inhibit ovulatory cycles but still decrease the risk for ovarian epithelial cancers
  • Polycystic ovarian syndrome (PCOS) decreases the number of ovulatory cycles but increases the risk for ovarian epithelial cancer.
Gonadotropins[1][204][205][206][207][208][209]
  • Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) and human chorionic gonadotropin stimulate ovarian epithelial cells proliferation
  • Resultant increased mitotic activity make cells more susceptible to mutations
  • Higher incidences of epthelial ovarian cancers in women taking infertility drugs in some studies
  • Polycystic ovarian syndrome (PCOS) and infertility increase the risk for ovarian epithelial cancers
  • Progesterone only oral contraceptives decrease the risk for ovarian epithelial cancers
  • Follicle-stimulating hormone (FSH) and luteinizing hormone (LH) are shown to increase cell proliferation in some studies
  • Up-regulation of Cox-1 and Cox-2 and resultant increase in PGE2 by follicle-stimulating hormone (FSH) and luteinizing hormone (LH) has been observed
  • Up-regulation of potential oncogenes in vitro such as EGFR, HER-2, and c-myc, cyclin G2, Meis-1, β-catenin, β-1 integrin, and IGF-1 by Follicle-stimulating hormone (FSH) receptor over-expression
  • Some studies suggest that infertility, rather than gonadotropin drugs treatment, increases the susceptibility to epithelial ovarian cancers
  • No concrete linkage of gonadotropins to malignant transformation of surface epithelial cells of ovaries
  • Proposed hypothesis of gonadotropin role in tumor cell growth and survival rather than origin
Hormonal influence[1][203][204][210][211][212]
  • Androgens confer greater risk to epithelial ovarian cancer while progesterone decreases the rik
  • Conditions that result in androgenic excess such as Polycystic ovarian syndrome (PCOS), hirsutism, and acne have been shown to increase the risk for epithelial ovarian cancer
  • Andorgens can stimulate cellular proliferation (androgens are are thought to be pre-dominant in ovarian inclusion cysts)
  • Progesterone only oral contraceptives decrease the risk for ovarian epithelial cancers, possibly through decreased androgens
  • No concrete linkage of androgens to malignant transformation of surface epithelial cells of ovaries
  • No evidence of androgens and their precursors affecting cancer cells growth
Inflammation[1][208][213][214]
  • Cytokines and inflammatory cells are involved in ovulation and repair and increase susceptibility to mutations and carcinogenesis
  • Non-steroidal anti-inflammatory drugs (NSAIDS) and Aminosalicylic acid (ASA) are thought to decrease the risk for epithelial ovarian cancer
  • Chemicals that cause inflammation such as talc and asbestos are shown to be associated with an increased risk for epithelial ovarian cancer
  • Inflammatory pathways and mediators have been observed in tumor pathogenesis and tumor micro-environment
  • No established linkage between talc and asbestos with pathogenesis of epithelial ovarian cancers in animal studies

Obesity: A Risk Factor for Epithelial Cancer

  • A British study comprising of 1.2 million women found that incidences of epithelial ovarian cancer were higher among women with BMI >30 as compared to women with normal BMI, with risk increasing with incremental increase in BMI. A meta-analysis conducted Olsen et al.also found an increase risk for epithelial ovarian cancer in obese women.[215][216][217]
  • It has been hypothesized that waist to hip ratio provides a better risk determination for epithelial ovarian cancer because of more accuracy in assessing true visceral fat deposition but remains to be validated.[215][218]
  • The time at which women develop obesity during their life may be a key factor for increased risk for epithelial ovarian cancer. Multiple studies indicate that increased BMI in adolescence and/or early adulthood may confer a greater risk for developing epithelial ovarian cancer.[215][219][220]
  • Another study postulates that duration and severity of obesity is also associated with increased risk for epithelial ovarian cancer and few others postulate that association of obesity with epithelial ovarian cancer is greater in premenopausal women than post-menopausal.[215][216][221]
  • Another meta-analysis demonstrated that obesity is associated with not only an increased risk for epithelial ovarian cancer but also with decrease in overall survival and ovarian-cancer specific survival. Another study also showed an increase in ovarian cancer- related mortality in obese women.[215][222][223]

Diabetes Mellitus and The Risk of Epithelial Ovarian Cancer

  • While conflicting data is present for association of diabetes mellitus and an increased risk for epithelial ovarian cancer, multiple studies, however, demonstrated diabetes as an independent risk factor for increased mortality in epithelial ovarian cancer.[215][224][225][226][227]
  • Findings in some studies indicate a greater risk for epithelial ovarian cancer in diabetic women while some suggest an increased risk only in pre-menopausal women, and some suggest no increase in risk for epithelial ovarian cancer at all.[215][224][225][226]

Metabolic Syndrome and the Risk of Epithelial Ovarian Cancer

  • The case for metabolic syndrome to be associated with an increased risk for epithelial ovarian cancer is similar to that of diabetes mellitus. There has been a fewer studies on association between metabolic syndrome and epithelial ovarian cancer and the results are conflicting with some found an increased risk for epithelial ovarian cancer in women with metabolic syndrome while some found no association.[215][228][229]
  • But an association of metabolic syndrome with increased ovarian cancer-related mortality was found in these studies. These studies however had limitation of lack of racial diversity because the study sample comprised only of Caucasian women.[215][229]

Pathogenesis of Epithelial Ovarian Cancer Associated with Metabolic Abnormalities

  • The work on mechanisms linking metabolic abnormalities to epithelial ovarian cancer is not yet complete and the way by which these abnormalities confer a greater risk for epithelial ovarian cancer is not well-understood but several theories have been put forward.
  • The most significant of these theories include role of cytokines and adipokines, immune cells, and aberrant signaling pathways in association with increased risk for epithelial ovarian cancer in women with metabolic derangement.

Cytokines and Adipokines

The role of cytokines and adipokines in epithelial ovarian cancer
Cytokines and adipokines Association with metabolic abnormalities Proposed mechanism in initiation and progression of epithelial ovarian cancer
Tissue necrosis factor-α[215][230][231][232]
  • Produced by immune cells (macrophages), tumor cells and fat cells
  • Shown to be elevated in obesity and diabetes mellitus
  • Promotes matrix metalloproteinases that contribute to carcinognesis and increased risk for tumor cell invasion and metastasis
  • Promotes tumor cells growth by acting as paracrine and autocrine growth factor
  • Promotes angiogenesis that contribute to tumor progression
  • Promotes cell survival
  • Promotes cell proliferation
  • Inhibits apoptosis
  • Acts to decrease adiponectin levels by decreasing its production
  • Promotes aromatase expression in adipose tissues
  • Promotes insulin resistance
  • Promotes inflammation
  • A positive correlation of tissue necrosis factor-α levels with tumor grade of epithelial ovarian cancer
  • Elevated levels shown to be associated with decreased overall survival
Leptin[215][233][234][235][236]
  • Produced by adipocytes
  • Shown to be elevated in obesity and produced by tumor cells
  • Leptin receptors expressed by tumor cells
  • Inhibits natural killer function by decreasing
    • toxicity towards tumor cells
    • perforin production
    • interferon-γ secretion
  • Promotes secretion of interleukin-6 and tissue necrosis factor-α by monocytes
  • Promotes tumor cells growth and invasion
  • Promotes resistance to apoptosis
  • Promotes tumor cells proliferation
  • Promotes expression of cyclin-D that increases tumor cells growth and survival
  • Promotes tumor cells migration
  • Shown to decrease progression-free survival in epithelial ovarian tumors
IL-6[215][235][237]
  • Produced by immune cells (macrophages), tumor cells and fat cells
  • Shown to be elevated in obesity and diabetes mellitus
  • Reactive oxygen species associated with an increased level of interlekin-6
  • Promotes angiogenesis
  • Associated with increased aromatase that leads to elevated levels of estrogen
  • Inhibits apoptosis by increasing expression of anti-apoptotic proteins
  • Promotes resistance to chemotherapy
  • Promotes inflammation
  • Associated with increased levels of C-reactive protein
  • Response prediction to bevacizumab therapy
C reactive protein (CRP)[215][232]
  • Produced by liver
  • Shown to be elevated in obesity and diabetes mellitus
  • Shown to be associated with an increased risk for developing epithelial ovarian cancer
Monocyte chemotactic protein-1 (MCP-1)[215][233][238]
  • Produced by ovarian tumor cells
  • Induced by hypoxia inducible factor (levels elevated in obesity)
  • Elevated levels observed in obesity
  • Promotes monocytes recruitment
  • Associated with increased density of tumor associated macrophages
  • May play a role in angiogenesis
Adiponectin[215][218][239][240]
  • Produced by mature fat cells
  • Shown to be decreased in obesity and diabetes mellitus
  • Anti-tumor effects lost/decreased in obesity and diabetes mellitus that include
  • insulin sensitivity
  • Inhibition of inflammation
  • inhibition of tumor growth
  • inhibition of angiogenesis
  • inhibition of tissue necrosis factor-α signaling

Immune Cells

  • Immune cells may have a pro or anti-tumor effect, depending on the cell type. Metabolic risk factors may alter these cell types and their functions to have a promoter effect in initiation and progression of epithelial ovarian tumors.[215]
  • The table below provides a short overview of possible role of immune cells in pathogenesis of epithelial ovarian tumors.[215]
The role of immune cells in epithelial ovarian cancer
Cell type Link with metabolic risk factors Possible role in pathogenesis
Dendritic cells[241][242][243]
  • Tumor stroma-derived factor 1 (SDF-1) recruits dendritic cells
  • Interleukin 10 by tumor cells leads to alteration in dendritic cells differentiation
  • These specific subtypes induced by tumor cells cytokines are less efficient in T-cells activation
  • Interact with programmed death-ligand 1 (PD-L1) to decrease T-cells effector function
Macrophages[238][244][245][246][247]
  • Two populations:
    • (1) M1 → classically activated tumor associated macrophages
    • (2) M2 → alternatively activated tumor associated macrophages
  • Studies indicate decreased M1 subpopulation in obese patients leading to ↓ M1/M2 ratio
  • Interferon gamma induce differentiation of macrophages into M1 subpopulation
  • Differentiation of macrophages into M2 subpopulation is possibly promoted by
    • transforming growth factor-beta
    • interleukin-4
    • interleukin-10
    • interleukin-13
    • colony stimulating factor-1
  • M1
    • inhibit tumor progression
    • promote inflammation through production of cytokines
    • cytotoxic to tumor cell
    • produce and release reactive oxygen species (ROS)
  • M2
    • promote tumor cells growth
    • promote angiogenesis
    • promote invasion and metastasis
    • promote tissue repair
    • inhibits immune system
    • produce C-C motif chemokine 22 (CCL22)
Natural killer cells[248][249]
  • Leptin may inhibit cytotoxic activity and interferon-γ production by natural killer cells
  • Mucin 16, Cell Surface Associated (MUC16) suppresses natural killer cells function
  • Increased natural killer cells activity in peripheral blood → increased progression free survival
  • Higher number of natural killer cells in peritoneal/pleural fluids → poor prognosis
B-cells[247][250][251]
  • Animal model studies indicate an up to threefold increase in B cells population in tumor cells in obese patients
  • Higher B cells tumor population associated with poorer prognosis
  • Promote angiogenesis
T-cells[248][252][253][254]
  • Interaction with programmed death-ligand 1 (PD-L1) inhibits CD4+ and CD8+ cells function
  • Transforming growth factor-β inhibits cytotoxic function of CD8+ cells
  • C-C motif chemokine 22 (CCL22) recruits regulatory T cells to the tumor
  • CD4+ cells
    • produce interleukin-17 that may have an anti-tumor role
  • CD8+ cells
    • increased number is associated with increased survival
  • Regulatory T-cells
    • release interleukin-10 and transforming growth factor-β that inhibit anti-tumor function of T-cells
    • inhibit cytotoxic function of CD8+ T cells

Hormones, Signaling Pathways in Pathogenesis of Epithelial Ovarian Cancer and Their Link to Metabolic Risk Factors

  • Hormones and signaling pathways that may play a role in pathogenesis of epithelial ovarian cancer with link to metabolic abnormalities are summarized below in table:[215]
Possible role of hormones and signal transduction pathways in relation to metabolic abnormalities
Hormone Link with metabolic risk factors Possible role in pathogenesis
Hypoxia inducible factor (HIF)[233][230][255][256]
  • Increased in obesity due to relative hypoxia
  • Promotes growth and survival by increased production/expression of
    • interleukin-6
    • tissue necrosis factor
    • monocyte chemoattractant protein-1 (MCP-1)
    • erythropoietin
    • vascular endothelial growth factor and vascular endothelial growth factor receptors
    • glucose transporters
    • glycolytic enzymes
  • Promotes inflammation through increased expression of cytokines and recruitment of macrophages
  • Leads to increased C-X-C chemokine receptor type 4 (CXCR-4) that leads to increased density of tumor associated macrophages
  • Promotes angiogenesis
  • Promotes pro-fibrotic pathway, resulting in altered extra-cellular matrix
Vascular endothelial growth factor (VEGF)[247][257]
  • Increased levels observed in obesity
  • Promotes angiogenesis
  • Promotes tumor cells growth
  • Promotes metastasis
Insulin-like growth factor 1 (IGF-1)[233][225][258][259]
  • Increased levels observed in obesity
  • Increased levels in diabetes mellitus patients
  • Promotes tumor cells growth
  • Promotes angiogenesis
  • Inhibits apoptosis
  • Decreases sex hormone binding globulin, leading to increased levels of biologically available estrogen
  • Associated with increased levels of hypoxia inducible factor (HIF)
  • Correlates negatively with survival
Estrogen[233][260][261]
  • Increased levels observed in obesity
  • Promotes tumor cells growth through action as a mitogen
  • Associated with increased expression of insulin-like growth factor 1 (IGF-1) receptors


References

  1. 1.0 1.1 1.2 1.3 1.4 1.5 Saad AF, Hu W, Sood AK (December 2010). "Microenvironment and pathogenesis of epithelial ovarian cancer". Horm Cancer. 1 (6): 277–90. doi:10.1007/s12672-010-0054-2. PMC 3199131. PMID 21761359.
  2. 2.00 2.01 2.02 2.03 2.04 2.05 2.06 2.07 2.08 2.09 2.10 2.11 2.12 2.13 2.14 2.15 2.16 2.17 2.18 2.19 2.20 2.21 Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC (April 2001). "Ovarian surface epithelium: biology, endocrinology, and pathology". Endocr. Rev. 22 (2): 255–88. doi:10.1210/edrv.22.2.0422. PMID 11294827.
  3. 3.0 3.1 Yoshinaga K, Hess DL, Hendrickx AG, Zamboni L (January 1988). "The development of the sexually indifferent gonad in the prosimian, Galago crassicaudatus crassicaudatus". Am. J. Anat. 181 (1): 89–105. doi:10.1002/aja.1001810110. PMID 3348150.
  4. 4.0 4.1 4.2 Siemens CH, Auersperg N (March 1988). "Serial propagation of human ovarian surface epithelium in tissue culture". J. Cell. Physiol. 134 (3): 347–56. doi:10.1002/jcp.1041340305. PMID 2450877.
  5. 5.0 5.1 5.2 Kruk PA, Uitto VJ, Firth JD, Dedhar S, Auersperg N (November 1994). "Reciprocal interactions between human ovarian surface epithelial cells and adjacent extracellular matrix". Exp. Cell Res. 215 (1): 97–108. doi:10.1006/excr.1994.1320. PMID 7525326.
  6. Davies BR, Worsley SD, Ponder BA (January 1998). "Expression of E-cadherin, alpha-catenin and beta-catenin in normal ovarian surface epithelium and epithelial ovarian cancers". Histopathology. 32 (1): 69–80. PMID 9522220.
  7. Sundfeldt K, Piontkewitz Y, Ivarsson K, Nilsson O, Hellberg P, Brännström M, Janson PO, Enerback S, Hedin L (June 1997). "E-cadherin expression in human epithelial ovarian cancer and normal ovary". Int. J. Cancer. 74 (3): 275–80. PMID 9221804.
  8. Osterholzer HO, Streibel EJ, Nicosia SV (August 1985). "Growth effects of protein hormones on cultured rabbit ovarian surface epithelial cells". Biol. Reprod. 33 (1): 247–58. PMID 3933584.
  9. Davies BR, Finnigan DS, Smith SK, Ponder BA (April 1999). "Administration of gonadotropins stimulates proliferation of normal mouse ovarian surface epithelium". Gynecol. Endocrinol. 13 (2): 75–81. PMID 10399050.
  10. Rodriguez GC, Berchuck A, Whitaker RS, Schlossman D, Clarke-Pearson DL, Bast RC (March 1991). "Epidermal growth factor receptor expression in normal ovarian epithelium and ovarian cancer. II. Relationship between receptor expression and response to epidermal growth factor". Am. J. Obstet. Gynecol. 164 (3): 745–50. PMID 2003535.
  11. Evangelou A, Jindal SK, Brown TJ, Letarte M (February 2000). "Down-regulation of transforming growth factor beta receptors by androgen in ovarian cancer cells". Cancer Res. 60 (4): 929–35. PMID 10706107.
  12. Kang SK, Choi KC, Tai CJ, Auersperg N, Leung PC (February 2001). "Estradiol regulates gonadotropin-releasing hormone (GnRH) and its receptor gene expression and antagonizes the growth inhibitory effects of GnRH in human ovarian surface epithelial and ovarian cancer cells". Endocrinology. 142 (2): 580–8. doi:10.1210/endo.142.2.7982. PMID 11159828.
  13. Liu Y, Lin L, Zarnegar R (September 1994). "Modulation of hepatocyte growth factor gene expression by estrogen in mouse ovary". Mol. Cell. Endocrinol. 104 (2): 173–81. PMID 7988745.
  14. Basilico C, Moscatelli D (1992). "The FGF family of growth factors and oncogenes". Adv. Cancer Res. 59: 115–65. PMID 1381547.
  15. Dabrow MB, Francesco MR, McBrearty FX, Caradonna S (October 1998). "The effects of platelet-derived growth factor and receptor on normal and neoplastic human ovarian surface epithelium". Gynecol. Oncol. 71 (1): 29–37. doi:10.1006/gyno.1998.5121. PMID 9784315.
  16. Wu S, Rodabaugh K, Martinez-Maza O, Watson JM, Silberstein DS, Boyer CM, Peters WP, Weinberg JB, Berek JS, Bast RC (March 1992). "Stimulation of ovarian tumor cell proliferation with monocyte products including interleukin-1, interleukin-6, and tumor necrosis factor-alpha". Am. J. Obstet. Gynecol. 166 (3): 997–1007. PMID 1550178.
  17. Wu S, Boyer CM, Whitaker RS, Berchuck A, Wiener JR, Weinberg JB, Bast RC (April 1993). "Tumor necrosis factor alpha as an autocrine and paracrine growth factor for ovarian cancer: monokine induction of tumor cell proliferation and tumor necrosis factor alpha expression". Cancer Res. 53 (8): 1939–44. PMID 8385577.
  18. Berchuck A, Rodriguez G, Olt G, Whitaker R, Boente MP, Arrick BA, Clarke-Pearson DL, Bast RC (February 1992). "Regulation of growth of normal ovarian epithelial cells and ovarian cancer cell lines by transforming growth factor-beta". Am. J. Obstet. Gynecol. 166 (2): 676–84. PMID 1536252.
  19. Parrott JA, Skinner MK (March 2000). "Expression and action of hepatocyte growth factor in human and bovine normal ovarian surface epithelium and ovarian cancer". Biol. Reprod. 62 (3): 491–500. PMID 10684788.
  20. Gulati R, Peluso JJ (May 1997). "Opposing actions of hepatocyte growth factor and basic fibroblast growth factor on cell contact, intracellular free calcium levels, and rat ovarian surface epithelial cell viability". Endocrinology. 138 (5): 1847–56. doi:10.1210/endo.138.5.5137. PMID 9112378.
  21. Ziltener HJ, Maines-Bandiera S, Schrader JW, Auersperg N (September 1993). "Secretion of bioactive interleukin-1, interleukin-6, and colony-stimulating factors by human ovarian surface epithelium". Biol. Reprod. 49 (3): 635–41. PMID 7691194.
  22. Marth C, Zeimet AG, Herold M, Brumm C, Windbichler G, Müller-Holzner E, Offner F, Feichtinger H, Zwierzina H, Daxenbichler G (September 1996). "Different effects of interferons, interleukin-1beta and tumor necrosis factor-alpha in normal (OSE) and malignant human ovarian epithelial cells". Int. J. Cancer. 67 (6): 826–30. doi:10.1002/(SICI)1097-0215(19960917)67:6<826::AID-IJC12>3.0.CO;2-#. PMID 8824555.
  23. 23.0 23.1 23.2 23.3 23.4 23.5 Devouassoux-Shisheboran M, Genestie C (January 2015). "Pathobiology of ovarian carcinomas". Chin J Cancer. 34 (1): 50–5. doi:10.5732/cjc.014.10273. PMC 4302089. PMID 25556618.
  24. 24.0 24.1 24.2 24.3 24.4 24.5 Lauchlan SC (July 1984). "Metaplasias and neoplasias of Müllerian epithelium". Histopathology. 8 (4): 543–57. PMID 6090303.
  25. 25.00 25.01 25.02 25.03 25.04 25.05 25.06 25.07 25.08 25.09 25.10 25.11 25.12 25.13 25.14 25.15 25.16 25.17 25.18 25.19 25.20 25.21 25.22 25.23 25.24 25.25 25.26 25.27 25.28 25.29 25.30 25.31 25.32 25.33 25.34 25.35 25.36 25.37 25.38 25.39 25.40 25.41 25.42 25.43 25.44 25.45 Kurman RJ, Shih I (July 2011). "Molecular pathogenesis and extraovarian origin of epithelial ovarian cancer--shifting the paradigm". Hum. Pathol. 42 (7): 918–31. doi:10.1016/j.humpath.2011.03.003. PMC 3148026. PMID 21683865. Vancouver style error: initials (help)
  26. 26.00 26.01 26.02 26.03 26.04 26.05 26.06 26.07 26.08 26.09 26.10 26.11 26.12 26.13 26.14 26.15 26.16 26.17 26.18 26.19 26.20 26.21 26.22 26.23 26.24 26.25 Dubeau L (December 2008). "The cell of origin of ovarian epithelial tumours". Lancet Oncol. 9 (12): 1191–7. doi:10.1016/S1470-2045(08)70308-5. PMC 4176875. PMID 19038766.
  27. 27.00 27.01 27.02 27.03 27.04 27.05 27.06 27.07 27.08 27.09 27.10 27.11 27.12 27.13 27.14 27.15 27.16 27.17 27.18 27.19 27.20 27.21 27.22 27.23 27.24 27.25 Kurman RJ, Shih I (April 2008). "Pathogenesis of ovarian cancer: lessons from morphology and molecular biology and their clinical implications". Int. J. Gynecol. Pathol. 27 (2): 151–60. doi:10.1097/PGP.0b013e318161e4f5. PMC 2794425. PMID 18317228. Vancouver style error: initials (help)
  28. 28.0 28.1 28.2 28.3 Riopel MA, Ronnett BM, Kurman RJ (June 1999). "Evaluation of diagnostic criteria and behavior of ovarian intestinal-type mucinous tumors: atypical proliferative (borderline) tumors and intraepithelial, microinvasive, invasive, and metastatic carcinomas". Am. J. Surg. Pathol. 23 (6): 617–35. PMID 10366144.
  29. 29.0 29.1 29.2 29.3 29.4 29.5 29.6 29.7 Kindelberger DW, Lee Y, Miron A, Hirsch MS, Feltmate C, Medeiros F, Callahan MJ, Garner EO, Gordon RW, Birch C, Berkowitz RS, Muto MG, Crum CP (February 2007). "Intraepithelial carcinoma of the fimbria and pelvic serous carcinoma: Evidence for a causal relationship". Am. J. Surg. Pathol. 31 (2): 161–9. doi:10.1097/01.pas.0000213335.40358.47. PMID 17255760.
  30. 30.0 30.1 30.2 Marquez RT, Baggerly KA, Patterson AP, Liu J, Broaddus R, Frumovitz M, Atkinson EN, Smith DI, Hartmann L, Fishman D, Berchuck A, Whitaker R, Gershenson DM, Mills GB, Bast RC, Lu KH (September 2005). "Patterns of gene expression in different histotypes of epithelial ovarian cancer correlate with those in normal fallopian tube, endometrium, and colon". Clin. Cancer Res. 11 (17): 6116–26. doi:10.1158/1078-0432.CCR-04-2509. PMID 16144910.
  31. 31.0 31.1 31.2 31.3 Callahan MJ, Crum CP, Medeiros F, Kindelberger DW, Elvin JA, Garber JE, Feltmate CM, Berkowitz RS, Muto MG (September 2007). "Primary fallopian tube malignancies in BRCA-positive women undergoing surgery for ovarian cancer risk reduction". J. Clin. Oncol. 25 (25): 3985–90. doi:10.1200/JCO.2007.12.2622. PMID 17761984.
  32. 32.0 32.1 32.2 32.3 Piek JM, van Diest PJ, Zweemer RP, Jansen JW, Poort-Keesom RJ, Menko FH, Gille JJ, Jongsma AP, Pals G, Kenemans P, Verheijen RH (November 2001). "Dysplastic changes in prophylactically removed Fallopian tubes of women predisposed to developing ovarian cancer". J. Pathol. 195 (4): 451–6. doi:10.1002/path.1000. PMID 11745677.
  33. Pothuri B, Leitao MM, Levine DA, Viale A, Olshen AB, Arroyo C, Bogomolniy F, Olvera N, Lin O, Soslow RA, Robson ME, Offit K, Barakat RR, Boyd J (April 2010). "Genetic analysis of the early natural history of epithelial ovarian carcinoma". PLoS ONE. 5 (4): e10358. doi:10.1371/journal.pone.0010358. PMC 2859950. PMID 20436685.
  34. 34.0 34.1 Devouassoux-Shisheboran M, Genestie C (January 2015). "Pathobiology of ovarian carcinomas". Chin J Cancer. 34 (1): 50–5. doi:10.5732/cjc.014.10273. PMC 4302089. PMID 25556618.
  35. 35.0 35.1 Veras E, Mao TL, Ayhan A, Ueda S, Lai H, Hayran M, Shih I, Kurman RJ (June 2009). "Cystic and adenofibromatous clear cell carcinomas of the ovary: distinctive tumors that differ in their pathogenesis and behavior: a clinicopathologic analysis of 122 cases". Am. J. Surg. Pathol. 33 (6): 844–53. doi:10.1097/PAS.0b013e31819c4271. PMID 19342944. Vancouver style error: initials (help)
  36. 36.0 36.1 Martin DC (1997). "Cancer and endometriosis: do we need to be concerned?". Semin. Reprod. Endocrinol. 15 (3): 319–24. doi:10.1055/s-2008-1068762. PMID 9383841.
  37. Piek JM, Verheijen RH, Kenemans P, Massuger LF, Bulten H, van Diest PJ (August 2003). "BRCA1/2-related ovarian cancers are of tubal origin: a hypothesis". Gynecol. Oncol. 90 (2): 491. PMID 12893227.
  38. Medeiros F, Muto MG, Lee Y, Elvin JA, Callahan MJ, Feltmate C, Garber JE, Cramer DW, Crum CP (February 2006). "The tubal fimbria is a preferred site for early adenocarcinoma in women with familial ovarian cancer syndrome". Am. J. Surg. Pathol. 30 (2): 230–6. PMID 16434898.
  39. Lee TY, Lee C, Choi WJ, Lee JY, Kim HY (July 2013). "Synchronous occurrence of primary malignant mixed müllerian tumor in ovary and uterus". Obstet Gynecol Sci. 56 (4): 269–72. doi:10.5468/ogs.2013.56.4.269. PMC 3784138. PMID 24328014.
  40. Gupta AJ, Singh M, Rani P, Jain S, Khurana N, Sahu L (2017). "Malignant mixed mullerian tumor of ovary-scrape cytology: Findings with review of literature". J Cytol. 34 (2): 125–126. doi:10.4103/0970-9371.203568. PMC 5398022. PMID 28469326.
  41. Piek JM, van Diest PJ, Zweemer RP, Kenemans P, Verheijen RH (September 2001). "Tubal ligation and risk of ovarian cancer". Lancet. 358 (9284): 844. doi:10.1016/S0140-6736(01)05992-X. PMID 11570411.
  42. Rosenblatt KA, Thomas DB (November 1996). "Reduced risk of ovarian cancer in women with a tubal ligation or hysterectomy. The World Health Organization Collaborative Study of Neoplasia and Steroid Contraceptives". Cancer Epidemiol. Biomarkers Prev. 5 (11): 933–5. PMID 8922304.
  43. 43.0 43.1 Vang R, Gown AM, Zhao C, Barry TS, Isacson C, Richardson MS, Ronnett BM (June 2007). "Ovarian mucinous tumors associated with mature cystic teratomas: morphologic and immunohistochemical analysis identifies a subset of potential teratomatous origin that shares features of lower gastrointestinal tract mucinous tumors more commonly encountered as secondary tumors in the ovary". Am. J. Surg. Pathol. 31 (6): 854–69. doi:10.1097/PAS.0b013e31802efb45. PMID 17527072.
  44. 44.0 44.1 Seidman JD, Khedmati F (November 2008). "Exploring the histogenesis of ovarian mucinous and transitional cell (Brenner) neoplasms and their relationship with Walthard cell nests: a study of 120 tumors". Arch. Pathol. Lab. Med. 132 (11): 1753–60. doi:10.1043/1543-2165-132.11.1753. PMID 18976011.
  45. 45.0 45.1 45.2 45.3 45.4 45.5 45.6 45.7 45.8 45.9 Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, Schilder RJ, Cortesi L (2015). "Hereditary ovarian cancer: not only BRCA 1 and 2 genes". Biomed Res Int. 2015: 341723. doi:10.1155/2015/341723. PMC 4449870. PMID 26075229.
  46. Devouassoux-Shisheboran M, Genestie C (January 2015). "Pathobiology of ovarian carcinomas". Chin J Cancer. 34 (1): 50–5. doi:10.5732/cjc.014.10273. PMC 4302089. PMID 25556618.
  47. 47.0 47.1 47.2 47.3 47.4 47.5 47.6 47.7 47.8 Lynch HT, Casey MJ, Snyder CL, Bewtra C, Lynch JF, Butts M, Godwin AK (April 2009). "Hereditary ovarian carcinoma: heterogeneity, molecular genetics, pathology, and management". Mol Oncol. 3 (2): 97–137. doi:10.1016/j.molonc.2009.02.004. PMID 19383374.
  48. 48.0 48.1 48.2 48.3 48.4 48.5 48.6 Neff RT, Senter L, Salani R (August 2017). "BRCA mutation in ovarian cancer: testing, implications and treatment considerations". Ther Adv Med Oncol. 9 (8): 519–531. doi:10.1177/1758834017714993. PMID 28794804.
  49. 49.0 49.1 49.2 49.3 49.4 49.5 Martín-López JV, Fishel R (June 2013). "The mechanism of mismatch repair and the functional analysis of mismatch repair defects in Lynch syndrome". Fam. Cancer. 12 (2): 159–68. doi:10.1007/s10689-013-9635-x. PMC 4235668. PMID 23572416.
  50. 50.00 50.01 50.02 50.03 50.04 50.05 50.06 50.07 50.08 50.09 50.10 50.11 50.12 50.13 50.14 50.15 50.16 50.17 50.18 50.19 Roy R, Chun J, Powell SN (December 2011). "BRCA1 and BRCA2: different roles in a common pathway of genome protection". Nat. Rev. Cancer. 12 (1): 68–78. doi:10.1038/nrc3181. PMC 4972490. PMID 22193408.
  51. Wu W, Koike A, Takeshita T, Ohta T (January 2008). "The ubiquitin E3 ligase activity of BRCA1 and its biological functions". Cell Div. 3: 1. doi:10.1186/1747-1028-3-1. PMC 2254412. PMID 18179693.
  52. 52.0 52.1 52.2 Thorslund T, McIlwraith MJ, Compton SA, Lekomtsev S, Petronczki M, Griffith JD, West SC (October 2010). "The breast cancer tumor suppressor BRCA2 promotes the specific targeting of RAD51 to single-stranded DNA". Nat. Struct. Mol. Biol. 17 (10): 1263–5. doi:10.1038/nsmb.1905. PMC 4041013. PMID 20729858.
  53. 53.0 53.1 53.2 Carreira A, Hilario J, Amitani I, Baskin RJ, Shivji MK, Venkitaraman AR, Kowalczykowski SC (March 2009). "The BRC repeats of BRCA2 modulate the DNA-binding selectivity of RAD51". Cell. 136 (6): 1032–43. doi:10.1016/j.cell.2009.02.019. PMC 2669112. PMID 19303847.
  54. 54.0 54.1 54.2 Sy SM, Huen MS, Chen J (April 2009). "PALB2 is an integral component of the BRCA complex required for homologous recombination repair". Proc. Natl. Acad. Sci. U.S.A. 106 (17): 7155–60. doi:10.1073/pnas.0811159106. PMC 2678481. PMID 19369211.
  55. 55.0 55.1 55.2 Xia B, Sheng Q, Nakanishi K, Ohashi A, Wu J, Christ N, Liu X, Jasin M, Couch FJ, Livingston DM (June 2006). "Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2". Mol. Cell. 22 (6): 719–29. doi:10.1016/j.molcel.2006.05.022. PMID 16793542.
  56. 56.0 56.1 56.2 Hsieh P, Yamane K (2008). "DNA mismatch repair: molecular mechanism, cancer, and ageing". Mech. Ageing Dev. 129 (7–8): 391–407. doi:10.1016/j.mad.2008.02.012. PMC 2574955. PMID 18406444.
  57. 57.0 57.1 57.2 57.3 Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, Schilder RJ, Cortesi L (2015). "Hereditary ovarian cancer: not only BRCA 1 and 2 genes". Biomed Res Int. 2015: 341723. doi:10.1155/2015/341723. PMC 4449870. PMID 26075229.
  58. 58.0 58.1 58.2 58.3 Miller M, Shirole N, Tian R, Pal D, Sordella R (2016). "The Evolution of TP53 Mutations: From Loss-of-Function to Separation-of-Function Mutants". J Cancer Biol Res. 4 (4). PMC 5298884. PMID 28191499.
  59. 59.0 59.1 59.2 59.3 Toss A, Tomasello C, Razzaboni E, Contu G, Grandi G, Cagnacci A, Schilder RJ, Cortesi L (2015). "Hereditary ovarian cancer: not only BRCA 1 and 2 genes". Biomed Res Int. 2015: 341723. doi:10.1155/2015/341723. PMC 4449870. PMID 26075229.
  60. Pantziarka P (2015). "Primed for cancer: Li Fraumeni Syndrome and the pre-cancerous niche". Ecancermedicalscience. 9: 541. doi:10.3332/ecancer.2015.541. PMC 4462886. PMID 26082798.
  61. D'Andrilli G, Giordano A, Bovicelli A (February 2008). "Epithelial ovarian cancer: the role of cell cycle genes in the different histotypes". Open Clin Cancer J. 2: 7–12. doi:10.2174/1874189400802010007. PMC 2490600. PMID 18665245.
  62. 62.0 62.1 62.2 62.3 62.4 Shih I, Kurman RJ (May 2004). "Ovarian tumorigenesis: a proposed model based on morphological and molecular genetic analysis". Am. J. Pathol. 164 (5): 1511–8. PMID 15111296. Vancouver style error: initials (help)
  63. 63.0 63.1 Rojas V, Hirshfield KM, Ganesan S, Rodriguez-Rodriguez L (December 2016). "Molecular Characterization of Epithelial Ovarian Cancer: Implications for Diagnosis and Treatment". Int J Mol Sci. 17 (12). doi:10.3390/ijms17122113. PMC 5187913. PMID 27983698.
  64. 64.0 64.1 McCluggage WG (August 2011). "Morphological subtypes of ovarian carcinoma: a review with emphasis on new developments and pathogenesis". Pathology. 43 (5): 420–32. doi:10.1097/PAT.0b013e328348a6e7. PMID 21716157.
  65. 65.0 65.1 Wiegand KC, Shah SP, Al-Agha OM, Zhao Y, Tse K, Zeng T, Senz J, McConechy MK, Anglesio MS, Kalloger SE, Yang W, Heravi-Moussavi A, Giuliany R, Chow C, Fee J, Zayed A, Prentice L, Melnyk N, Turashvili G, Delaney AD, Madore J, Yip S, McPherson AW, Ha G, Bell L, Fereday S, Tam A, Galletta L, Tonin PN, Provencher D, Miller D, Jones SJ, Moore RA, Morin GB, Oloumi A, Boyd N, Aparicio SA, Shih I, Mes-Masson AM, Bowtell DD, Hirst M, Gilks B, Marra MA, Huntsman DG (October 2010). "ARID1A mutations in endometriosis-associated ovarian carcinomas". N. Engl. J. Med. 363 (16): 1532–43. doi:10.1056/NEJMoa1008433. PMC 2976679. PMID 20942669. Vancouver style error: initials (help)
  66. Ahmed AA, Etemadmoghadam D, Temple J, Lynch AG, Riad M, Sharma R, Stewart C, Fereday S, Caldas C, Defazio A, Bowtell D, Brenton JD (May 2010). "Driver mutations in TP53 are ubiquitous in high grade serous carcinoma of the ovary". J. Pathol. 221 (1): 49–56. doi:10.1002/path.2696. PMC 3262968. PMID 20229506.
  67. 67.0 67.1 Senturk E, Cohen S, Dottino PR, Martignetti JA (November 2010). "A critical re-appraisal of BRCA1 methylation studies in ovarian cancer". Gynecol. Oncol. 119 (2): 376–83. doi:10.1016/j.ygyno.2010.07.026. PMID 20797776.
  68. 68.0 68.1 Burks RT, Sherman ME, Kurman RJ (November 1996). "Micropapillary serous carcinoma of the ovary. A distinctive low-grade carcinoma related to serous borderline tumors". Am. J. Surg. Pathol. 20 (11): 1319–30. PMID 8898836.
  69. Ho CL, Kurman RJ, Dehari R, Wang TL, Shih I (October 2004). "Mutations of BRAF and KRAS precede the development of ovarian serous borderline tumors". Cancer Res. 64 (19): 6915–8. doi:10.1158/0008-5472.CAN-04-2067. PMID 15466181. Vancouver style error: initials (help)
  70. Singer G, Oldt R, Cohen Y, Wang BG, Sidransky D, Kurman RJ, Shih I (March 2003). "Mutations in BRAF and KRAS characterize the development of low-grade ovarian serous carcinoma". J. Natl. Cancer Inst. 95 (6): 484–6. PMID 12644542. Vancouver style error: initials (help)
  71. Mayr D, Hirschmann A, Löhrs U, Diebold J (December 2006). "KRAS and BRAF mutations in ovarian tumors: a comprehensive study of invasive carcinomas, borderline tumors and extraovarian implants". Gynecol. Oncol. 103 (3): 883–7. doi:10.1016/j.ygyno.2006.05.029. PMID 16806438.
  72. Hsu CY, Bristow R, Cha MS, Wang BG, Ho CL, Kurman RJ, Wang TL, Shih I (October 2004). "Characterization of active mitogen-activated protein kinase in ovarian serous carcinomas". Clin. Cancer Res. 10 (19): 6432–6. doi:10.1158/1078-0432.CCR-04-0893. PMID 15475429. Vancouver style error: initials (help)
  73. May T, Virtanen C, Sharma M, Milea A, Begley H, Rosen B, Murphy KJ, Brown TJ, Shaw PA (April 2010). "Low malignant potential tumors with micropapillary features are molecularly similar to low-grade serous carcinoma of the ovary". Gynecol. Oncol. 117 (1): 9–17. doi:10.1016/j.ygyno.2010.01.006. PMID 20117829.
  74. Nakayama K, Nakayama N, Jinawath N, Salani R, Kurman RJ, Shih I, Wang TL (June 2007). "Amplicon profiles in ovarian serous carcinomas". Int. J. Cancer. 120 (12): 2613–7. doi:10.1002/ijc.22609. PMID 17351921. Vancouver style error: initials (help)
  75. 75.0 75.1 Kroeger PT, Drapkin R (February 2017). "Pathogenesis and heterogeneity of ovarian cancer". Curr. Opin. Obstet. Gynecol. 29 (1): 26–34. doi:10.1097/GCO.0000000000000340. PMC 5201412. PMID 27898521.
  76. Karst AM, Drapkin R (2010). "Ovarian cancer pathogenesis: a model in evolution". J Oncol. 2010: 932371. doi:10.1155/2010/932371. PMC 2739011. PMID 19746182.
  77. Jones S, Wang TL, Shih I, Mao TL, Nakayama K, Roden R, Glas R, Slamon D, Diaz LA, Vogelstein B, Kinzler KW, Velculescu VE, Papadopoulos N (October 2010). "Frequent mutations of chromatin remodeling gene ARID1A in ovarian clear cell carcinoma". Science. 330 (6001): 228–31. doi:10.1126/science.1196333. PMC 3076894. PMID 20826764. Vancouver style error: initials (help)
  78. Campbell IG, Russell SE, Choong DY, Montgomery KG, Ciavarella ML, Hooi CS, Cristiano BE, Pearson RB, Phillips WA (November 2004). "Mutation of the PIK3CA gene in ovarian and breast cancer". Cancer Res. 64 (21): 7678–81. doi:10.1158/0008-5472.CAN-04-2933. PMID 15520168.
  79. 79.0 79.1 Sato N, Tsunoda H, Nishida M, Morishita Y, Takimoto Y, Kubo T, Noguchi M (December 2000). "Loss of heterozygosity on 10q23.3 and mutation of the tumor suppressor gene PTEN in benign endometrial cyst of the ovary: possible sequence progression from benign endometrial cyst to endometrioid carcinoma and clear cell carcinoma of the ovary". Cancer Res. 60 (24): 7052–6. PMID 11156411.
  80. Chandler RL, Damrauer JS, Raab JR, Schisler JC, Wilkerson MD, Didion JP, Starmer J, Serber D, Yee D, Xiong J, Darr DB, Pardo-Manuel de Villena F, Kim WY, Magnuson T (January 2015). "Coexistent ARID1A-PIK3CA mutations promote ovarian clear-cell tumorigenesis through pro-tumorigenic inflammatory cytokine signalling". Nat Commun. 6: 6118. doi:10.1038/ncomms7118. PMC 4308813. PMID 25625625.
  81. 81.0 81.1 Obata K, Morland SJ, Watson RH, Hitchcock A, Chenevix-Trench G, Thomas EJ, Campbell IG (May 1998). "Frequent PTEN/MMAC mutations in endometrioid but not serous or mucinous epithelial ovarian tumors". Cancer Res. 58 (10): 2095–7. PMID 9605750.
  82. 82.0 82.1 Catasús L, Bussaglia E, Rodrguez I, Gallardo A, Pons C, Irving JA, Prat J (November 2004). "Molecular genetic alterations in endometrioid carcinomas of the ovary: similar frequency of beta-catenin abnormalities but lower rate of microsatellite instability and PTEN alterations than in uterine endometrioid carcinomas". Hum. Pathol. 35 (11): 1360–8. doi:10.1016/j.humpath.2004.07.019. PMID 15668893.
  83. 83.0 83.1 83.2 Wu R, Hendrix-Lucas N, Kuick R, Zhai Y, Schwartz DR, Akyol A, Hanash S, Misek DE, Katabuchi H, Williams BO, Fearon ER, Cho KR (April 2007). "Mouse model of human ovarian endometrioid adenocarcinoma based on somatic defects in the Wnt/beta-catenin and PI3K/Pten signaling pathways". Cancer Cell. 11 (4): 321–33. doi:10.1016/j.ccr.2007.02.016. PMID 17418409.
  84. Ichikawa Y, Nishida M, Suzuki H, Yoshida S, Tsunoda H, Kubo T, Uchida K, Miwa M (January 1994). "Mutation of K-ras protooncogene is associated with histological subtypes in human mucinous ovarian tumors". Cancer Res. 54 (1): 33–5. PMID 8261457.
  85. Gemignani ML, Schlaerth AC, Bogomolniy F, Barakat RR, Lin O, Soslow R, Venkatraman E, Boyd J (August 2003). "Role of KRAS and BRAF gene mutations in mucinous ovarian carcinoma". Gynecol. Oncol. 90 (2): 378–81. PMID 12893203.
  86. 86.0 86.1 Wee P, Wang Z (May 2017). "Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways". Cancers (Basel). 9 (5). doi:10.3390/cancers9050052. PMC 5447962. PMID 28513565.
  87. 87.0 87.1 Iqbal N, Iqbal N (2014). "Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications". Mol Biol Int. 2014: 852748. doi:10.1155/2014/852748. PMC 4170925. PMID 25276427.
  88. Zan L, Wu H, Jiang J, Zhao S, Song Y, Teng G, Li H, Jia Y, Zhou M, Zhang X, Qi J, Wang J (July 2011). "Temporal profile of Src, SSeCKS, and angiogenic factors after focal cerebral ischemia: correlations with angiogenesis and cerebral edema". Neurochem. Int. 58 (8): 872–9. doi:10.1016/j.neuint.2011.02.014. PMC 3100427. PMID 21334414.
  89. Reinecke JB, Katafiasz D, Naslavsky N, Caplan S (April 2014). "Regulation of Src trafficking and activation by the endocytic regulatory proteins MICAL-L1 and EHD1". J. Cell. Sci. 127 (Pt 8): 1684–98. doi:10.1242/jcs.133892. PMC 3986674. PMID 24481818.
  90. Dwyer AR, Greenland EL, Pixley FJ (June 2017). "Promotion of Tumor Invasion by Tumor-Associated Macrophages: The Role of CSF-1-Activated Phosphatidylinositol 3 Kinase and Src Family Kinase Motility Signaling". Cancers (Basel). 9 (6). doi:10.3390/cancers9060068. PMC 5483887. PMID 28629162.
  91. Abraham D, Zins K, Sioud M, Lucas T, Schäfer R, Stanley ER, Aharinejad S (March 2010). "Stromal cell-derived CSF-1 blockade prolongs xenograft survival of CSF-1-negative neuroblastoma". Int. J. Cancer. 126 (6): 1339–52. doi:10.1002/ijc.24859. PMC 3222589. PMID 19711348.
  92. Laron Z (October 2001). "Insulin-like growth factor 1 (IGF-1): a growth hormone". MP, Mol. Pathol. 54 (5): 311–6. PMC 1187088. PMID 11577173.
  93. Weroha SJ, Haluska P (June 2012). "The insulin-like growth factor system in cancer". Endocrinol. Metab. Clin. North Am. 41 (2): 335–50, vi. doi:10.1016/j.ecl.2012.04.014. PMC 3614012. PMID 22682634.
  94. Lukanova A, Lundin E, Toniolo P, Micheli A, Akhmedkhanov A, Rinaldi S, Muti P, Lenner P, Biessy C, Krogh V, Zeleniuch-Jacquotte A, Berrino F, Hallmans G, Riboli E, Kaaks R (October 2002). "Circulating levels of insulin-like growth factor-I and risk of ovarian cancer". Int. J. Cancer. 101 (6): 549–54. doi:10.1002/ijc.10613. PMID 12237896.
  95. Prior IA, Lewis PD, Mattos C (May 2012). "A comprehensive survey of Ras mutations in cancer". Cancer Res. 72 (10): 2457–67. doi:10.1158/0008-5472.CAN-11-2612. PMC 3354961. PMID 22589270.
  96. Franklin WA, Haney J, Sugita M, Bemis L, Jimeno A, Messersmith WA (January 2010). "KRAS mutation: comparison of testing methods and tissue sampling techniques in colon cancer". J Mol Diagn. 12 (1): 43–50. doi:10.2353/jmoldx.2010.080131. PMC 2797717. PMID 20007845.
  97. Estep AL, Palmer C, McCormick F, Rauen KA (December 2007). "Mutation analysis of BRAF, MEK1 and MEK2 in 15 ovarian cancer cell lines: implications for therapy". PLoS ONE. 2 (12): e1279. doi:10.1371/journal.pone.0001279. PMC 2093994. PMID 18060073.
  98. Grisham RN, Iyer G, Garg K, Delair D, Hyman DM, Zhou Q, Iasonos A, Berger MF, Dao F, Spriggs DR, Levine DA, Aghajanian C, Solit DB (February 2013). "BRAF mutation is associated with early stage disease and improved outcome in patients with low-grade serous ovarian cancer". Cancer. 119 (3): 548–554. doi:10.1002/cncr.27782. PMC 3961140. PMID 22930283.
  99. Alsina-Sanchís E, Figueras A, Lahiguera A, Gil-Martín M, Pardo B, Piulats JM, Martí L, Ponce J, Matias-Guiu X, Vidal A, Villanueva A, Viñals F (July 2017). "TGFβ Controls Ovarian Cancer Cell Proliferation". Int J Mol Sci. 18 (8). doi:10.3390/ijms18081658. PMC 5578048. PMID 28758950.
  100. Principe DR, Doll JA, Bauer J, Jung B, Munshi HG, Bartholin L, Pasche B, Lee C, Grippo PJ (February 2014). "TGF-β: duality of function between tumor prevention and carcinogenesis". J. Natl. Cancer Inst. 106 (2): djt369. doi:10.1093/jnci/djt369. PMC 3952197. PMID 24511106.
  101. Bierie B, Moses HL (February 2010). "Transforming growth factor beta (TGF-beta) and inflammation in cancer". Cytokine Growth Factor Rev. 21 (1): 49–59. doi:10.1016/j.cytogfr.2009.11.008. PMC 2834863. PMID 20018551.
  102. Miller DM, Thomas SD, Islam A, Muench D, Sedoris K (October 2012). "c-Myc and cancer metabolism". Clin. Cancer Res. 18 (20): 5546–53. doi:10.1158/1078-0432.CCR-12-0977. PMC 3505847. PMID 23071356.
  103. Dang CV (March 2012). "MYC on the path to cancer". Cell. 149 (1): 22–35. doi:10.1016/j.cell.2012.03.003. PMC 3345192. PMID 22464321.
  104. Aughey GN, Grice SJ, Liu JL (February 2016). "The Interplay between Myc and CTP Synthase in Drosophila". PLoS Genet. 12 (2): e1005867. doi:10.1371/journal.pgen.1005867. PMC 4759343. PMID 26889675.
  105. Neumeister P, Pixley FJ, Xiong Y, Xie H, Wu K, Ashton A, Cammer M, Chan A, Symons M, Stanley ER, Pestell RG (May 2003). "Cyclin D1 governs adhesion and motility of macrophages". Mol. Biol. Cell. 14 (5): 2005–15. doi:10.1091/mbc.02-07-0102. PMC 165093. PMID 12802071.
  106. Dong P, Zhang C, Parker BT, You L, Mathey-Prevot B (2018). "Cyclin D/CDK4/6 activity controls G1 length in mammalian cells". PLoS ONE. 13 (1): e0185637. doi:10.1371/journal.pone.0185637. PMC 5757913. PMID 29309421.
  107. Khleif SN, DeGregori J, Yee CL, Otterson GA, Kaye FJ, Nevins JR, Howley PM (April 1996). "Inhibition of cyclin D-CDK4/CDK6 activity is associated with an E2F-mediated induction of cyclin kinase inhibitor activity". Proc. Natl. Acad. Sci. U.S.A. 93 (9): 4350–4. PMC 39540. PMID 8633069.
  108. Honda R, Lowe ED, Dubinina E, Skamnaki V, Cook A, Brown NR, Johnson LN (February 2005). "The structure of cyclin E1/CDK2: implications for CDK2 activation and CDK2-independent roles". EMBO J. 24 (3): 452–63. doi:10.1038/sj.emboj.7600554. PMC 548659. PMID 15660127.
  109. Choudhary GS, Tat TT, Misra S, Hill BT, Smith MR, Almasan A, Mazumder S (July 2015). "Cyclin E/Cdk2-dependent phosphorylation of Mcl-1 determines its stability and cellular sensitivity to BH3 mimetics". Oncotarget. 6 (19): 16912–25. doi:10.18632/oncotarget.4857. PMC 4627281. PMID 26219338.
  110. Won KA, Reed SI (August 1996). "Activation of cyclin E/CDK2 is coupled to site-specific autophosphorylation and ubiquitin-dependent degradation of cyclin E". EMBO J. 15 (16): 4182–93. PMC 452142. PMID 8861947.
  111. Sun X, Zhangyuan G, Shi L, Wang Y, Sun B, Ding Q (May 2017). "Prognostic and clinicopathological significance of cyclin B expression in patients with breast cancer: A meta-analysis". Medicine (Baltimore). 96 (19): e6860. doi:10.1097/MD.0000000000006860. PMC 5428614. PMID 28489780.
  112. Huang Y, Sramkoski RM, Jacobberger JW (2013). "The kinetics of G2 and M transitions regulated by B cyclins". PLoS ONE. 8 (12): e80861. doi:10.1371/journal.pone.0080861. PMC 3851588. PMID 24324638.
  113. Lindqvist A, van Zon W, Karlsson Rosenthal C, Wolthuis RM (May 2007). "Cyclin B1-Cdk1 activation continues after centrosome separation to control mitotic progression". PLoS Biol. 5 (5): e123. doi:10.1371/journal.pbio.0050123. PMC 1858714. PMID 17472438.
  114. Yoon N, Yoon G, Park CK, Kim HS (October 2016). "Stromal p16 expression is significantly increased in malignant ovarian neoplasms". Oncotarget. 7 (40): 64665–64673. doi:10.18632/oncotarget.11660. PMC 5323106. PMID 27572321.
  115. Sano T, Oyama T, Kashiwabara K, Fukuda T, Nakajima T (December 1998). "Expression status of p16 protein is associated with human papillomavirus oncogenic potential in cervical and genital lesions". Am. J. Pathol. 153 (6): 1741–8. doi:10.1016/S0002-9440(10)65689-1. PMC 1866324. PMID 9846965.
  116. Felix AS, Sherman ME, Hewitt SM, Gunja MZ, Yang HP, Cora RL, Boudreau V, Ylaya K, Lissowska J, Brinton LA, Wentzensen N (2015). "Cell-cycle protein expression in a population-based study of ovarian and endometrial cancers". Front Oncol. 5: 25. doi:10.3389/fonc.2015.00025. PMC 4321403. PMID 25709969.
  117. Lee J, Kim SS (November 2009). "The function of p27 KIP1 during tumor development". Exp. Mol. Med. 41 (11): 765–71. doi:10.3858/emm.2009.41.11.102. PMC 2788730. PMID 19887899.
  118. Roy S, Singh RP, Agarwal C, Siriwardana S, Sclafani R, Agarwal R (June 2008). "Downregulation of both p21/Cip1 and p27/Kip1 produces a more aggressive prostate cancer phenotype". Cell Cycle. 7 (12): 1828–35. doi:10.4161/cc.7.12.6024. PMC 2744498. PMID 18583941.
  119. Miskimins WK, Wang G, Hawkinson M, Miskimins R (August 2001). "Control of cyclin-dependent kinase inhibitor p27 expression by cap-independent translation". Mol. Cell. Biol. 21 (15): 4960–7. doi:10.1128/MCB.21.15.4960-4967.2001. PMC 87223. PMID 11438653.
  120. Abbas T, Dutta A (June 2009). "p21 in cancer: intricate networks and multiple activities". Nat. Rev. Cancer. 9 (6): 400–14. doi:10.1038/nrc2657. PMC 2722839. PMID 19440234.
  121. Dash BC, El-Deiry WS (April 2005). "Phosphorylation of p21 in G2/M promotes cyclin B-Cdc2 kinase activity". Mol. Cell. Biol. 25 (8): 3364–87. doi:10.1128/MCB.25.8.3364-3387.2005. PMC 1069593. PMID 15798220.
  122. Shi Y, Zou M, Farid NR, al-Sedairy ST (November 1996). "Evidence of gene deletion of p21 (WAF1/CIP1), a cyclin-dependent protein kinase inhibitor, in thyroid carcinomas". Br. J. Cancer. 74 (9): 1336–41. PMC 2074763. PMID 8912526.
  123. Lawrence T (December 2009). "The nuclear factor NF-kappaB pathway in inflammation". Cold Spring Harb Perspect Biol. 1 (6): a001651. doi:10.1101/cshperspect.a001651. PMC 2882124. PMID 20457564.
  124. Yang G, Xiao X, Rosen DG, Cheng X, Wu X, Chang B, Liu G, Xue F, Mercado-Uribe I, Chiao P, Du X, Liu J (April 2011). "The biphasic role of NF-kappaB in progression and chemoresistance of ovarian cancer". Clin. Cancer Res. 17 (8): 2181–94. doi:10.1158/1078-0432.CCR-10-3265. PMC 3152795. PMID 21339307.
  125. Charbonneau B, Block MS, Bamlet WR, Vierkant RA, Kalli KR, Fogarty Z, Rider DN, Sellers TA, Tworoger SS, Poole E, Risch HA, Salvesen HB, Kiemeney LA, Baglietto L, Giles GG, Severi G, Trabert B, Wentzensen N, Chenevix-Trench G, Whittemore AS, Sieh W, Chang-Claude J, Bandera EV, Orlow I, Terry K, Goodman MT, Thompson PJ, Cook LS, Rossing MA, Ness RB, Narod SA, Kupryjanczyk J, Lu K, Butzow R, Dörk T, Pejovic T, Campbell I, Le ND, Bunker CH, Bogdanova N, Runnebaum IB, Eccles D, Paul J, Wu AH, Gayther SA, Hogdall E, Heitz F, Kaye SB, Karlan BY, Anton-Culver H, Gronwald J, Hogdall CK, Lambrechts D, Fasching PA, Menon U, Schildkraut J, Pearce CL, Levine DA, Kjaer SK, Cramer D, Flanagan JM, Phelan CM, Brown R, Massuger LF, Song H, Doherty JA, Krakstad C, Liang D, Odunsi K, Berchuck A, Jensen A, Lubinski J, Nevanlinna H, Bean YT, Lurie G, Ziogas A, Walsh C, Despierre E, Brinton L, Hein A, Rudolph A, Dansonka-Mieszkowska A, Olson SH, Harter P, Tyrer J, Vitonis AF, Brooks-Wilson A, Aben KK, Pike MC, Ramus SJ, Wik E, Cybulski C, Lin J, Sucheston L, Edwards R, McGuire V, Lester J, du Bois A, Lundvall L, Wang-Gohrke S, Szafron LM, Lambrechts S, Yang H, Beckmann MW, Pelttari LM, Van Altena AM, van den Berg D, Halle MK, Gentry-Maharaj A, Schwaab I, Chandran U, Menkiszak J, Ekici AB, Wilkens LR, Leminen A, Modugno F, Friel G, Rothstein JH, Vergote I, Garcia-Closas M, Hildebrandt MA, Sobiczewski P, Kelemen LE, Pharoah PD, Moysich K, Knutson KL, Cunningham JM, Fridley BL, Goode EL (February 2014). "Risk of ovarian cancer and the NF-κB pathway: genetic association with IL1A and TNFSF10". Cancer Res. 74 (3): 852–61. doi:10.1158/0008-5472.CAN-13-1051. PMC 3946482. PMID 24272484.
  126. Badgwell DB, Lu Z, Le K, Gao F, Yang M, Suh GK, Bao JJ, Das P, Andreeff M, Chen W, Yu Y, Ahmed AA, S-L Liao W, Bast RC (January 2012). "The tumor-suppressor gene ARHI (DIRAS3) suppresses ovarian cancer cell migration through inhibition of the Stat3 and FAK/Rho signaling pathways". Oncogene. 31 (1): 68–79. doi:10.1038/onc.2011.213. PMC 3170676. PMID 21643014.
  127. Yu Y, Xu F, Peng H, Fang X, Zhao S, Li Y, Cuevas B, Kuo WL, Gray JW, Siciliano M, Mills GB, Bast RC (January 1999). "NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas". Proc. Natl. Acad. Sci. U.S.A. 96 (1): 214–9. PMC 15119. PMID 9874798.
  128. Albanese C, Johnson J, Watanabe G, Eklund N, Vu D, Arnold A, Pestell RG (October 1995). "Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions". J. Biol. Chem. 270 (40): 23589–97. PMID 7559524.
  129. Dobbin ZC, Landen CN (April 2013). "The importance of the PI3K/AKT/MTOR pathway in the progression of ovarian cancer". Int J Mol Sci. 14 (4): 8213–27. doi:10.3390/ijms14048213. PMC 3645739. PMID 23591839.
  130. Hemmings BA, Restuccia DF (September 2012). "PI3K-PKB/Akt pathway". Cold Spring Harb Perspect Biol. 4 (9): a011189. doi:10.1101/cshperspect.a011189. PMC 3428770. PMID 22952397.
  131. Nitulescu GM, Van De Venter M, Nitulescu G, Ungurianu A, Juzenas P, Peng Q, Olaru OT, Grădinaru D, Tsatsakis A, Tsoukalas D, Spandidos DA, Margina D (December 2018). "The Akt pathway in oncology therapy and beyond (Review)". Int. J. Oncol. 53 (6): 2319–2331. doi:10.3892/ijo.2018.4597. PMC 6203150. PMID 30334567.
  132. Shi Y, Paluch BE, Wang X, Jiang X (October 2012). "PTEN at a glance". J. Cell. Sci. 125 (Pt 20): 4687–92. doi:10.1242/jcs.093765. PMC 3517091. PMID 23223894.
  133. Tanwar PS, Mohapatra G, Chiang S, Engler DA, Zhang L, Kaneko-Tarui T, Ohguchi Y, Birrer MJ, Teixeira JM (March 2014). "Loss of LKB1 and PTEN tumor suppressor genes in the ovarian surface epithelium induces papillary serous ovarian cancer". Carcinogenesis. 35 (3): 546–53. doi:10.1093/carcin/bgt357. PMC 3941742. PMID 24170201.
  134. Hopkins BD, Parsons RE (November 2014). "Molecular pathways: intercellular PTEN and the potential of PTEN restoration therapy". Clin. Cancer Res. 20 (21): 5379–83. doi:10.1158/1078-0432.CCR-13-2661. PMC 4362520. PMID 25361917.
  135. Zilfou JT, Lowe SW (November 2009). "Tumor suppressive functions of p53". Cold Spring Harb Perspect Biol. 1 (5): a001883. doi:10.1101/cshperspect.a001883. PMC 2773645. PMID 20066118.
  136. Ozaki T, Nakagawara A (March 2011). "Role of p53 in Cell Death and Human Cancers". Cancers (Basel). 3 (1): 994–1013. doi:10.3390/cancers3010994. PMC 3756401. PMID 24212651.
  137. Zhang Y, Cao L, Nguyen D, Lu H (December 2016). "TP53 mutations in epithelial ovarian cancer". Transl Cancer Res. 5 (6): 650–663. doi:10.21037/tcr.2016.08.40. PMC 6320227. PMID 30613473.
  138. 138.0 138.1 Chen S, Iversen ES, Friebel T, Finkelstein D, Weber BL, Eisen A, Peterson LE, Schildkraut JM, Isaacs C, Peshkin BN, Corio C, Leondaridis L, Tomlinson G, Dutson D, Kerber R, Amos CI, Strong LC, Berry DA, Euhus DM, Parmigiani G (February 2006). "Characterization of BRCA1 and BRCA2 mutations in a large United States sample". J. Clin. Oncol. 24 (6): 863–71. doi:10.1200/JCO.2005.03.6772. PMC 2323978. PMID 16484695.
  139. Vymetalkova VP, Slyskova J, Korenkova V, Bielik L, Langerova L, Prochazka P, Rejhova A, Schwarzova L, Pardini B, Naccarati A, Vodicka P (January 2014). "Molecular characteristics of mismatch repair genes in sporadic colorectal tumors in Czech patients". BMC Med. Genet. 15: 17. doi:10.1186/1471-2350-15-17. PMC 3913626. PMID 24484585.
  140. Murphy MA, Wentzensen N (October 2011). "Frequency of mismatch repair deficiency in ovarian cancer: a systematic review This article is a US Government work and, as such, is in the public domain of the United States of America". Int. J. Cancer. 129 (8): 1914–22. doi:10.1002/ijc.25835. PMC 3107885. PMID 21140452.
  141. Heinen CD (February 2016). "Mismatch repair defects and Lynch syndrome: The role of the basic scientist in the battle against cancer". DNA Repair (Amst.). 38: 127–34. doi:10.1016/j.dnarep.2015.11.025. PMC 4740212. PMID 26710976.
  142. Peter ME, Hadji A, Murmann AE, Brockway S, Putzbach W, Pattanayak A, Ceppi P (April 2015). "The role of CD95 and CD95 ligand in cancer". Cell Death Differ. 22 (4): 549–59. doi:10.1038/cdd.2015.3. PMC 4356349. PMID 25656654.
  143. Fraser M, Leung B, Jahani-Asl A, Yan X, Thompson WE, Tsang BK (October 2003). "Chemoresistance in human ovarian cancer: the role of apoptotic regulators". Reprod. Biol. Endocrinol. 1: 66. doi:10.1186/1477-7827-1-66. PMC 270001. PMID 14609433.
  144. Lowin B, Hahne M, Mattmann C, Tschopp J (August 1994). "Cytolytic T-cell cytotoxicity is mediated through perforin and Fas lytic pathways". Nature. 370 (6491): 650–2. doi:10.1038/370650a0. PMID 7520535.
  145. Morandi F, Rizzo R, Fainardi E, Rouas-Freiss N, Pistoia V (2016). "Recent Advances in Our Understanding of HLA-G Biology: Lessons from a Wide Spectrum of Human Diseases". J Immunol Res. 2016: 4326495. doi:10.1155/2016/4326495. PMC 5019910. PMID 27652273.
  146. Lin A, Yan WH (November 2015). "Human Leukocyte Antigen-G (HLA-G) Expression in Cancers: Roles in Immune Evasion, Metastasis and Target for Therapy". Mol. Med. 21 (1): 782–791. doi:10.2119/molmed.2015.00083. PMC 4749493. PMID 26322846.
  147. Sheu JJ, Shih I (December 2007). "Clinical and biological significance of HLA-G expression in ovarian cancer". Semin. Cancer Biol. 17 (6): 436–43. doi:10.1016/j.semcancer.2007.06.012. PMC 2151836. PMID 17681474. Vancouver style error: initials (help)
  148. Lee YK, Chung HH, Kim JW, Song YS, Park NH (2015). "Expression of phosphorylated Akt and hTERT is associated with prognosis of epithelial ovarian carcinoma". Int J Clin Exp Pathol. 8 (11): 14971–6. PMC 4713616. PMID 26823830.
  149. Ramlee MK, Wang J, Toh WX, Li S (August 2016). "Transcription Regulation of the Human Telomerase Reverse Transcriptase (hTERT) Gene". Genes (Basel). 7 (8). doi:10.3390/genes7080050. PMC 4999838. PMID 27548225.
  150. Leão R, Apolónio JD, Lee D, Figueiredo A, Tabori U, Castelo-Branco P (March 2018). "Mechanisms of human telomerase reverse transcriptase (hTERT) regulation: clinical impacts in cancer". J. Biomed. Sci. 25 (1): 22. doi:10.1186/s12929-018-0422-8. PMC 5846307. PMID 29526163.
  151. Masoumi Moghaddam S, Amini A, Morris DL, Pourgholami MH (June 2012). "Significance of vascular endothelial growth factor in growth and peritoneal dissemination of ovarian cancer". Cancer Metastasis Rev. 31 (1–2): 143–62. doi:10.1007/s10555-011-9337-5. PMC 3350632. PMID 22101807.
  152. Goel HL, Mercurio AM (December 2013). "VEGF targets the tumour cell". Nat. Rev. Cancer. 13 (12): 871–82. doi:10.1038/nrc3627. PMC 4011842. PMID 24263190.
  153. Ohta Y, Shridhar V, Bright RK, Kalemkerian GP, Du W, Carbone M, Watanabe Y, Pass HI (September 1999). "VEGF and VEGF type C play an important role in angiogenesis and lymphangiogenesis in human malignant mesothelioma tumours". Br. J. Cancer. 81 (1): 54–61. doi:10.1038/sj.bjc.6690650. PMC 2374345. PMID 10487612.
  154. David JM, Dominguez C, Hamilton DH, Palena C (June 2016). "The IL-8/IL-8R Axis: A Double Agent in Tumor Immune Resistance". Vaccines (Basel). 4 (3). doi:10.3390/vaccines4030022. PMC 5041016. PMID 27348007.
  155. Yung MM, Tang HW, Cai PC, Leung TH, Ngu SF, Chan KK, Xu D, Yang H, Ngan HY, Chan DW (2018). "GRO-α and IL-8 enhance ovarian cancer metastatic potential via the CXCR2-mediated TAK1/NFκB signaling cascade". Theranostics. 8 (5): 1270–1285. doi:10.7150/thno.22536. PMC 5835935. PMID 29507619.
  156. Escudero-Lourdes C, Wu T, Camarillo JM, Gandolfi AJ (January 2012). "Interleukin-8 (IL-8) over-production and autocrine cell activation are key factors in monomethylarsonous acid [MMA(III)]-induced malignant transformation of urothelial cells". Toxicol. Appl. Pharmacol. 258 (1): 10–8. doi:10.1016/j.taap.2011.10.002. PMC 3254786. PMID 22015448.
  157. Park JE, Son AI, Zhou R (July 2013). "Roles of EphA2 in Development and Disease". Genes (Basel). 4 (3): 334–57. doi:10.3390/genes4030334. PMC 3924825. PMID 24705208.
  158. Dunne PD, Dasgupta S, Blayney JK, McArt DG, Redmond KL, Weir JA, Bradley CA, Sasazuki T, Shirasawa S, Wang T, Srivastava S, Ong CW, Arthur K, Salto-Tellez M, Wilson RH, Johnston PG, Van Schaeybroeck S (January 2016). "EphA2 Expression Is a Key Driver of Migration and Invasion and a Poor Prognostic Marker in Colorectal Cancer". Clin. Cancer Res. 22 (1): 230–242. doi:10.1158/1078-0432.CCR-15-0603. PMC 4694030. PMID 26283684.
  159. Lu C, Shahzad MM, Wang H, Landen CN, Kim SW, Allen J, Nick AM, Jennings N, Kinch MS, Bar-Eli M, Sood AK (July 2008). "EphA2 overexpression promotes ovarian cancer growth". Cancer Biol. Ther. 7 (7): 1098–103. PMC 2705979. PMID 18443431.
  160. Page-McCaw A, Ewald AJ, Werb Z (March 2007). "Matrix metalloproteinases and the regulation of tissue remodelling". Nat. Rev. Mol. Cell Biol. 8 (3): 221–33. doi:10.1038/nrm2125. PMC 2760082. PMID 17318226.
  161. Caley MP, Martins VL, O'Toole EA (April 2015). "Metalloproteinases and Wound Healing". Adv Wound Care (New Rochelle). 4 (4): 225–234. doi:10.1089/wound.2014.0581. PMC 4397992. PMID 25945285.
  162. Al-Alem L, Curry TE (August 2015). "Ovarian cancer: involvement of the matrix metalloproteinases". Reproduction. 150 (2): R55–64. doi:10.1530/REP-14-0546. PMC 4955511. PMID 25918438.
  163. Cai WJ, Li MB, Wu X, Wu S, Zhu W, Chen D, Luo M, Eitenmüller I, Kampmann A, Schaper J, Schaper W (February 2009). "Activation of the integrins alpha 5beta 1 and alpha v beta 3 and focal adhesion kinase (FAK) during arteriogenesis". Mol. Cell. Biochem. 322 (1–2): 161–9. doi:10.1007/s11010-008-9953-8. PMC 2758386. PMID 18998200.
  164. Liu Z, Wang F, Chen X (2008). "Integrin alpha(v)beta(3)-Targeted Cancer Therapy". Drug Dev. Res. 69 (6): 329–339. doi:10.1002/ddr.20265. PMC 2901818. PMID 20628538.
  165. Shaw SK, Schreiber CL, Roland FM, Battles PM, Brennan SP, Padanilam SJ, Smith BD (May 2018). "High expression of integrin αvβ3 enables uptake of targeted fluorescent probes into ovarian cancer cells and tumors". Bioorg. Med. Chem. 26 (8): 2085–2091. doi:10.1016/j.bmc.2018.03.007. PMC 5963687. PMID 29548784.
  166. Shen Y, Schaller MD (August 1999). "Focal adhesion targeting: the critical determinant of FAK regulation and substrate phosphorylation". Mol. Biol. Cell. 10 (8): 2507–18. doi:10.1091/mbc.10.8.2507. PMC 25482. PMID 10436008.
  167. Zhao X, Guan JL (July 2011). "Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis". Adv. Drug Deliv. Rev. 63 (8): 610–5. doi:10.1016/j.addr.2010.11.001. PMC 3132829. PMID 21118706.
  168. Li M, Hong LI, Liao M, Guo G (August 2015). "Expression and clinical significance of focal adhesion kinase and adrenomedullin in epithelial ovarian cancer". Oncol Lett. 10 (2): 1003–1007. doi:10.3892/ol.2015.3278. PMC 4508992. PMID 26622614.
  169. Dong LL, Liu L, Ma CH, Li JS, Du C, Xu S, Han LH, Li L, Wang XW (June 2012). "E-cadherin promotes proliferation of human ovarian cancer cells in vitro via activating MEK/ERK pathway". Acta Pharmacol. Sin. 33 (6): 817–22. doi:10.1038/aps.2012.30. PMC 4010376. PMID 22543706.
  170. Pećina-Slaus N (October 2003). "Tumor suppressor gene E-cadherin and its role in normal and malignant cells". Cancer Cell Int. 3 (1): 17. doi:10.1186/1475-2867-3-17. PMC 270068. PMID 14613514.
  171. Petrova YI, Schecterson L, Gumbiner BM (November 2016). "Roles for E-cadherin cell surface regulation in cancer". Mol. Biol. Cell. 27 (21): 3233–3244. doi:10.1091/mbc.E16-01-0058. PMC 5170857. PMID 27582386.
  172. Fan S, Wang J, Yuan R, Ma Y, Meng Q, Erdos MR, Pestell RG, Yuan F, Auborn KJ, Goldberg ID, Rosen EM (May 1999). "BRCA1 inhibition of estrogen receptor signaling in transfected cells". Science. 284 (5418): 1354–6. PMID 10334989.
  173. Hamada J, Nakata D, Nakae D, Kobayashi Y, Akai H, Konishi Y, Okada F, Shibata T, Hosokawa M, Moriuchi T (February 2001). "Increased oxidative DNA damage in mammary tumor cells by continuous epidermal growth factor stimulation". J. Natl. Cancer Inst. 93 (3): 214–9. PMID 11158190.
  174. Famorca-Tran J, Roux G (2015). "The Consequences of a BRCA Mutation in Women". J Adv Pract Oncol. 6 (3): 194–210. PMC 4625626. PMID 26557407.
  175. 175.0 175.1 175.2 Sehgal R, Sheahan K, O'Connell PR, Hanly AM, Martin ST, Winter DC (June 2014). "Lynch syndrome: an updated review". Genes (Basel). 5 (3): 497–507. doi:10.3390/genes5030497. PMC 4198913. PMID 24978665.
  176. Hampel H, Frankel WL, Martin E, Arnold M, Khanduja K, Kuebler P, Clendenning M, Sotamaa K, Prior T, Westman JA, Panescu J, Fix D, Lockman J, LaJeunesse J, Comeras I, de la Chapelle A (December 2008). "Feasibility of screening for Lynch syndrome among patients with colorectal cancer". J. Clin. Oncol. 26 (35): 5783–8. doi:10.1200/JCO.2008.17.5950. PMC 2645108. PMID 18809606.
  177. Peltomäki P (July 2016). "Update on Lynch syndrome genomics". Fam. Cancer. 15 (3): 385–93. doi:10.1007/s10689-016-9882-8. PMC 4901089. PMID 26873718.
  178. Tabori U, Baskin B, Shago M, Alon N, Taylor MD, Ray PN, Bouffet E, Malkin D, Hawkins C (March 2010). "Universal poor survival in children with medulloblastoma harboring somatic TP53 mutations". J. Clin. Oncol. 28 (8): 1345–50. doi:10.1200/JCO.2009.23.5952. PMID 20142599.
  179. Levine AJ, Momand J, Finlay CA (June 1991). "The p53 tumour suppressor gene". Nature. 351 (6326): 453–6. doi:10.1038/351453a0. PMID 2046748.
  180. 180.0 180.1 180.2 Shulman LP, Dungan JS (2010). "Cancer genetics: risks and mechanisms of cancer in women with inherited susceptibility to epithelial ovarian cancer". Cancer Treat. Res. 156: 69–85. doi:10.1007/978-1-4419-6518-9_6. PMC 3086477. PMID 20811826.
  181. Stratton JF, Pharoah P, Smith SK, Easton D, Ponder BA (May 1998). "A systematic review and meta-analysis of family history and risk of ovarian cancer". Br J Obstet Gynaecol. 105 (5): 493–9. PMID 9637117.
  182. Lachlan KL, Lucassen AM, Bunyan D, Temple IK (September 2007). "Cowden syndrome and Bannayan Riley Ruvalcaba syndrome represent one condition with variable expression and age-related penetrance: results of a clinical study of PTEN mutation carriers". J. Med. Genet. 44 (9): 579–85. doi:10.1136/jmg.2007.049981. PMC 2597943. PMID 17526800.
  183. Kalin A, Merideth MA, Regier DS, Blumenthal GM, Dennis PA, Stratton P (February 2013). "Management of reproductive health in Cowden syndrome complicated by endometrial polyps and breast cancer". Obstet Gynecol. 121 (2 Pt 2 Suppl 1): 461–4. doi:http://10 1097/AOG.0b013e318270444f Check |doi= value (help). PMC 3799979. PMID 23344409.
  184. Loveday C, Turnbull C, Ramsay E, Hughes D, Ruark E, Frankum JR, Bowden G, Kalmyrzaev B, Warren-Perry M, Snape K, Adlard JW, Barwell J, Berg J, Brady AF, Brewer C, Brice G, Chapman C, Cook J, Davidson R, Donaldson A, Douglas F, Greenhalgh L, Henderson A, Izatt L, Kumar A, Lalloo F, Miedzybrodzka Z, Morrison PJ, Paterson J, Porteous M, Rogers MT, Shanley S, Walker L, Eccles D, Evans DG, Renwick A, Seal S, Lord CJ, Ashworth A, Reis-Filho JS, Antoniou AC, Rahman N (August 2011). "Germline mutations in RAD51D confer susceptibility to ovarian cancer". Nat. Genet. 43 (9): 879–882. doi:10.1038/ng.893. PMC 4845885. PMID 21822267.
  185. Meindl A, Hellebrand H, Wiek C, Erven V, Wappenschmidt B, Niederacher D, Freund M, Lichtner P, Hartmann L, Schaal H, Ramser J, Honisch E, Kubisch C, Wichmann HE, Kast K, Deissler H, Engel C, Müller-Myhsok B, Neveling K, Kiechle M, Mathew CG, Schindler D, Schmutzler RK, Hanenberg H (May 2010). "Germline mutations in breast and ovarian cancer pedigrees establish RAD51C as a human cancer susceptibility gene". Nat. Genet. 42 (5): 410–4. doi:10.1038/ng.569. PMID 20400964.
  186. Casadei S, Norquist BM, Walsh T, Stray S, Mandell JB, Lee MK, Stamatoyannopoulos JA, King MC (March 2011). "Contribution of inherited mutations in the BRCA2-interacting protein PALB2 to familial breast cancer". Cancer Res. 71 (6): 2222–9. doi:10.1158/0008-5472.CAN-10-3958. PMC 3059378. PMID 21285249.
  187. Poumpouridou N, Kroupis C (December 2011). "Hereditary breast cancer: beyond BRCA genetic analysis; PALB2 emerges". Clin. Chem. Lab. Med. 50 (3): 423–34. doi:10.1515/cclm-2011-0840. PMID 22505525.
  188. "CHEK2 gene - Genetics Home Reference - NIH".
  189. Cai Z, Chehab NH, Pavletich NP (September 2009). "Structure and activation mechanism of the CHK2 DNA damage checkpoint kinase". Mol. Cell. 35 (6): 818–29. doi:10.1016/j.molcel.2009.09.007. PMID 19782031.
  190. Meijers-Heijboer H, van den Ouweland A, Klijn J, Wasielewski M, de Snoo A, Oldenburg R, Hollestelle A, Houben M, Crepin E, van Veghel-Plandsoen M, Elstrodt F, van Duijn C, Bartels C, Meijers C, Schutte M, McGuffog L, Thompson D, Easton D, Sodha N, Seal S, Barfoot R, Mangion J, Chang-Claude J, Eccles D, Eeles R, Evans DG, Houlston R, Murday V, Narod S, Peretz T, Peto J, Phelan C, Zhang HX, Szabo C, Devilee P, Goldgar D, Futreal PA, Nathanson KL, Weber B, Rahman N, Stratton MR (May 2002). "Low-penetrance susceptibility to breast cancer due to CHEK2(*)1100delC in noncarriers of BRCA1 or BRCA2 mutations". Nat. Genet. 31 (1): 55–9. doi:10.1038/ng879. PMID 11967536.
  191. 191.0 191.1 Stracker TH, Petrini JH (February 2011). "The MRE11 complex: starting from the ends". Nat. Rev. Mol. Cell Biol. 12 (2): 90–103. doi:10.1038/nrm3047. PMC 3905242. PMID 21252998.
  192. Lamarche BJ, Orazio NI, Weitzman MD (September 2010). "The MRN complex in double-strand break repair and telomere maintenance". FEBS Lett. 584 (17): 3682–95. doi:10.1016/j.febslet.2010.07.029. PMC 2946096. PMID 20655309.
  193. Heikkinen K, Karppinen SM, Soini Y, Mäkinen M, Winqvist R (December 2003). "Mutation screening of Mre11 complex genes: indication of RAD50 involvement in breast and ovarian cancer susceptibility". J. Med. Genet. 40 (12): e131. PMC 1735331. PMID 14684699.
  194. Westermark UK, Reyngold M, Olshen AB, Baer R, Jasin M, Moynahan ME (November 2003). "BARD1 participates with BRCA1 in homology-directed repair of chromosome breaks". Mol. Cell. Biol. 23 (21): 7926–36. PMID 14560035.
  195. Brzovic PS, Rajagopal P, Hoyt DW, King MC, Klevit RE (October 2001). "Structure of a BRCA1-BARD1 heterodimeric RING-RING complex". Nat. Struct. Biol. 8 (10): 833–7. doi:10.1038/nsb1001-833. PMID 11573085.
  196. Klonowska K, Ratajska M, Czubak K, Kuzniacka A, Brozek I, Koczkowska M, Sniadecki M, Debniak J, Wydra D, Balut M, Stukan M, Zmienko A, Nowakowska B, Irminger-Finger I, Limon J, Kozlowski P (May 2015). "Analysis of large mutations in BARD1 in patients with breast and/or ovarian cancer: the Polish population as an example". Sci Rep. 5: 10424. doi:10.1038/srep10424. PMID 25994375.
  197. [++++https://ghr.nlm.nih.gov/gene/BRIP1 "BRIP1 gene - Genetics Home Reference - NIH"] Check |url= value (help).
  198. 198.0 198.1 Ring KL, Garcia C, Thomas MH, Modesitt SC (November 2017). "Current and future role of genetic screening in gynecologic malignancies". Am. J. Obstet. Gynecol. 217 (5): 512–521. doi:10.1016/j.ajog.2017.04.011. PMID 28411145.
  199. Weber-Lassalle N, Hauke J, Ramser J, Richters L, Groß E, Blümcke B, Gehrig A, Kahlert AK, Müller CR, Hackmann K, Honisch E, Weber-Lassalle K, Niederacher D, Borde J, Thiele H, Ernst C, Altmüller J, Neidhardt G, Nürnberg P, Klaschik K, Schroeder C, Platzer K, Volk AE, Wang-Gohrke S, Just W, Auber B, Kubisch C, Schmidt G, Horvath J, Wappenschmidt B, Engel C, Arnold N, Dworniczak B, Rhiem K, Meindl A, Schmutzler RK, Hahnen E (January 2018). "BRIP1 loss-of-function mutations confer high risk for familial ovarian cancer, but not familial breast cancer". Breast Cancer Res. 20 (1): 7. doi:10.1186/s13058-018-0935-9. PMC 5784717. PMID 29368626.
  200. Riman T, Dickman PW, Nilsson S, Correia N, Nordlinder H, Magnusson CM, Persson IR (August 2002). "Risk factors for invasive epithelial ovarian cancer: results from a Swedish case-control study". Am. J. Epidemiol. 156 (4): 363–73. PMID 12181107.
  201. Gwinn ML, Lee NC, Rhodes PH, Layde PM, Rubin GL (1990). "Pregnancy, breast feeding, and oral contraceptives and the risk of epithelial ovarian cancer". J Clin Epidemiol. 43 (6): 559–68. PMID 2348208.
  202. Nasca PC, Greenwald P, Chorost S, Richart R, Caputo T (May 1984). "An epidemiologic case-control study of ovarian cancer and reproductive factors". Am. J. Epidemiol. 119 (5): 705–13. PMID 6539067.
  203. 203.0 203.1 Risch HA (December 1998). "Hormonal etiology of epithelial ovarian cancer, with a hypothesis concerning the role of androgens and progesterone". J. Natl. Cancer Inst. 90 (23): 1774–86. PMID 9839517.
  204. 204.0 204.1 204.2 Schildkraut JM, Schwingl PJ, Bastos E, Evanoff A, Hughes C (October 1996). "Epithelial ovarian cancer risk among women with polycystic ovary syndrome". Obstet Gynecol. 88 (4 Pt 1): 554–9. PMID 8841217.
  205. Choi KC, Kang SK, Tai CJ, Auersperg N, Leung PC (May 2002). "Follicle-stimulating hormone activates mitogen-activated protein kinase in preneoplastic and neoplastic ovarian surface epithelial cells". J. Clin. Endocrinol. Metab. 87 (5): 2245–53. doi:10.1210/jcem.87.5.8506. PMID 11994371.
  206. Lau MT, Wong AS, Leung PC (July 2010). "Gonadotropins induce tumor cell migration and invasion by increasing cyclooxygenases expression and prostaglandin E(2) production in human ovarian cancer cells". Endocrinology. 151 (7): 2985–93. doi:10.1210/en.2009-1318. PMID 20392831.
  207. Choi JH, Choi KC, Auersperg N, Leung PC (November 2004). "Overexpression of follicle-stimulating hormone receptor activates oncogenic pathways in preneoplastic ovarian surface epithelial cells". J. Clin. Endocrinol. Metab. 89 (11): 5508–16. doi:10.1210/jc.2004-0044. PMID 15531506.
  208. 208.0 208.1 Ness RB, Cottreau C (September 1999). "Possible role of ovarian epithelial inflammation in ovarian cancer". J. Natl. Cancer Inst. 91 (17): 1459–67. PMID 10469746.
  209. Zheng W, Lu JJ, Luo F, Zheng Y, Feng Y, Felix JC, Lauchlan SC, Pike MC (January 2000). "Ovarian epithelial tumor growth promotion by follicle-stimulating hormone and inhibition of the effect by luteinizing hormone". Gynecol. Oncol. 76 (1): 80–8. doi:10.1006/gyno.1999.5628. PMID 10620446. Vancouver style error: initials (help)
  210. Rosenberg L, Palmer JR, Zauber AG, Warshauer ME, Lewis JL, Strom BL, Harlap S, Shapiro S (April 1994). "A case-control study of oral contraceptive use and invasive epithelial ovarian cancer". Am. J. Epidemiol. 139 (7): 654–61. PMID 8166126.
  211. Edmondson RJ, Monaghan JM, Davies BR (March 2002). "The human ovarian surface epithelium is an androgen responsive tissue". Br. J. Cancer. 86 (6): 879–85. doi:10.1038/sj.bjc.6600154. PMC 2364138. PMID 11953818.
  212. Seeger H, Wallwiener D, Mueck AO (2006). "Is there a protective role of progestogens on the proliferation of human ovarian cancer cells in the presence of growth factors?". Eur. J. Gynaecol. Oncol. 27 (2): 139–41. PMID 16620055.
  213. Altinoz MA, Korkmaz R (2004). "NF-kappaB, macrophage migration inhibitory factor and cyclooxygenase-inhibitions as likely mechanisms behind the acetaminophen- and NSAID-prevention of the ovarian cancer". Neoplasma. 51 (4): 239–47. PMID 15254653.
  214. Heller DS, Westhoff C, Gordon RE, Katz N (May 1996). "The relationship between perineal cosmetic talc usage and ovarian talc particle burden". Am. J. Obstet. Gynecol. 174 (5): 1507–10. PMID 9065120.
  215. 215.00 215.01 215.02 215.03 215.04 215.05 215.06 215.07 215.08 215.09 215.10 215.11 215.12 215.13 215.14 215.15 215.16 215.17 Craig ER, Londoño AI, Norian LA, Arend RC (December 2016). "Metabolic risk factors and mechanisms of disease in epithelial ovarian cancer: A review". Gynecol. Oncol. 143 (3): 674–683. doi:10.1016/j.ygyno.2016.10.005. PMC 5689410. PMID 27751590.
  216. 216.0 216.1 Reeves GK, Pirie K, Beral V, Green J, Spencer E, Bull D (December 2007). "Cancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study". BMJ. 335 (7630): 1134. doi:10.1136/bmj.39367.495995.AE. PMC 2099519. PMID 17986716.
  217. Olsen CM, Green AC, Whiteman DC, Sadeghi S, Kolahdooz F, Webb PM (March 2007). "Obesity and the risk of epithelial ovarian cancer: a systematic review and meta-analysis". Eur. J. Cancer. 43 (4): 690–709. doi:10.1016/j.ejca.2006.11.010. PMID 17223544.
  218. 218.0 218.1 Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M (February 2008). "Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies". Lancet. 371 (9612): 569–78. doi:10.1016/S0140-6736(08)60269-X. PMID 18280327.
  219. Lubin F, Chetrit A, Freedman LS, Alfandary E, Fishler Y, Nitzan H, Zultan A, Modan B (January 2003). "Body mass index at age 18 years and during adult life and ovarian cancer risk". Am. J. Epidemiol. 157 (2): 113–20. PMID 12522018.
  220. Engeland A, Tretli S, Bjørge T (August 2003). "Height, body mass index, and ovarian cancer: a follow-up of 1.1 million Norwegian women". J. Natl. Cancer Inst. 95 (16): 1244–8. PMID 12928351.
  221. Arnold M, Jiang L, Stefanick ML, Johnson KC, Lane DS, LeBlanc ES, Prentice R, Rohan TE, Snively BM, Vitolins M, Zaslavsky O, Soerjomataram I, Anton-Culver H (August 2016). "Duration of Adulthood Overweight, Obesity, and Cancer Risk in the Women's Health Initiative: A Longitudinal Study from the United States". PLoS Med. 13 (8): e1002081. doi:10.1371/journal.pmed.1002081. PMC 4987008. PMID 27529652.
  222. Protani MM, Nagle CM, Webb PM (July 2012). "Obesity and ovarian cancer survival: a systematic review and meta-analysis". Cancer Prev Res (Phila). 5 (7): 901–10. doi:10.1158/1940-6207.CAPR-12-0048. PMID 22609763.
  223. Nagle CM, Dixon SC, Jensen A, Kjaer SK, Modugno F, deFazio A, Fereday S, Hung J, Johnatty SE, Fasching PA, Beckmann MW, Lambrechts D, Vergote I, Van Nieuwenhuysen E, Lambrechts S, Risch HA, Rossing MA, Doherty JA, Wicklund KG, Chang-Claude J, Goodman MT, Ness RB, Moysich K, Heitz F, du Bois A, Harter P, Schwaab I, Matsuo K, Hosono S, Goode EL, Vierkant RA, Larson MC, Fridley BL, Høgdall C, Schildkraut JM, Weber RP, Cramer DW, Terry KL, Bandera EV, Paddock L, Rodriguez-Rodriguez L, Wentzensen N, Yang HP, Brinton LA, Lissowska J, Høgdall E, Lundvall L, Whittemore A, McGuire V, Sieh W, Rothstein J, Sutphen R, Anton-Culver H, Ziogas A, Pearce CL, Wu AH, Webb PM (September 2015). "Obesity and survival among women with ovarian cancer: results from the Ovarian Cancer Association Consortium". Br. J. Cancer. 113 (5): 817–26. doi:10.1038/bjc.2015.245. PMC 4559823. PMID 26151456.
  224. 224.0 224.1 Lee JY, Jeon I, Kim JW, Song YS, Yoon JM, Park SM (March 2013). "Diabetes mellitus and ovarian cancer risk: a systematic review and meta-analysis of observational studies". Int. J. Gynecol. Cancer. 23 (3): 402–12. doi:10.1097/IGC.0b013e31828189b2. PMID 23354371.
  225. 225.0 225.1 225.2 Vrachnis N, Iavazzo C, Iliodromiti Z, Sifakis S, Alexandrou A, Siristatidis C, Grigoriadis C, Botsis D, Creatsas G (February 2016). "Diabetes mellitus and gynecologic cancer: molecular mechanisms, epidemiological, clinical and prognostic perspectives". Arch. Gynecol. Obstet. 293 (2): 239–46. doi:10.1007/s00404-015-3858-z. PMID 26338721.
  226. 226.0 226.1 Chen HF, Chang YH, Ko MC, Li CY (September 2014). "A large scale population-based cohort study on the risk of ovarian neoplasm in patients with type 2 diabetes mellitus". Gynecol. Oncol. 134 (3): 576–80. doi:10.1016/j.ygyno.2014.07.001. PMID 25014539.
  227. Bakhru A, Buckanovich RJ, Griggs JJ (April 2011). "The impact of diabetes on survival in women with ovarian cancer". Gynecol. Oncol. 121 (1): 106–11. doi:10.1016/j.ygyno.2010.12.329. PMID 21236474.
  228. Esposito K, Chiodini P, Colao A, Lenzi A, Giugliano D (November 2012). "Metabolic syndrome and risk of cancer: a systematic review and meta-analysis". Diabetes Care. 35 (11): 2402–11. doi:10.2337/dc12-0336. PMC 3476894. PMID 23093685.
  229. 229.0 229.1 Bjørge T, Lukanova A, Tretli S, Manjer J, Ulmer H, Stocks T, Selmer R, Nagel G, Almquist M, Concin H, Hallmans G, Jonsson H, Häggström C, Stattin P, Engeland A (December 2011). "Metabolic risk factors and ovarian cancer in the Metabolic Syndrome and Cancer project". Int J Epidemiol. 40 (6): 1667–77. doi:10.1093/ije/dyr130. PMID 21984693.
  230. 230.0 230.1 Aggarwal BB, Shishodia S, Sandur SK, Pandey MK, Sethi G (November 2006). "Inflammation and cancer: how hot is the link?". Biochem. Pharmacol. 72 (11): 1605–21. doi:10.1016/j.bcp.2006.06.029. PMID 16889756.
  231. Naylor MS, Stamp GW, Foulkes WD, Eccles D, Balkwill FR (May 1993). "Tumor necrosis factor and its receptors in human ovarian cancer. Potential role in disease progression". J. Clin. Invest. 91 (5): 2194–206. doi:10.1172/JCI116446. PMC 288222. PMID 8387543.
  232. 232.0 232.1 Braun S, Bitton-Worms K, LeRoith D (2011). "The link between the metabolic syndrome and cancer". Int. J. Biol. Sci. 7 (7): 1003–15. PMC 3164150. PMID 21912508.
  233. 233.0 233.1 233.2 233.3 233.4 Park J, Morley TS, Kim M, Clegg DJ, Scherer PE (August 2014). "Obesity and cancer--mechanisms underlying tumour progression and recurrence". Nat Rev Endocrinol. 10 (8): 455–465. doi:10.1038/nrendo.2014.94. PMC 4374431. PMID 24935119.
  234. Uddin S, Bu R, Ahmed M, Abubaker J, Al-Dayel F, Bavi P, Al-Kuraya KS (September 2009). "Overexpression of leptin receptor predicts an unfavorable outcome in Middle Eastern ovarian cancer". Mol. Cancer. 8: 74. doi:10.1186/1476-4598-8-74. PMC 2754986. PMID 19765303.
  235. 235.0 235.1 Chen C, Chang YC, Lan MS, Breslin M (March 2013). "Leptin stimulates ovarian cancer cell growth and inhibits apoptosis by increasing cyclin D1 and Mcl-1 expression via the activation of the MEK/ERK1/2 and PI3K/Akt signaling pathways". Int. J. Oncol. 42 (3): 1113–9. doi:10.3892/ijo.2013.1789. PMID 23354006.
  236. Kato S, Abarzua-Catalan L, Trigo C, Delpiano A, Sanhueza C, García K, Ibañez C, Hormazábal K, Diaz D, Brañes J, Castellón E, Bravo E, Owen G, Cuello MA (August 2015). "Leptin stimulates migration and invasion and maintains cancer stem-like properties in ovarian cancer cells: an explanation for poor outcomes in obese women". Oncotarget. 6 (25): 21100–19. doi:10.18632/oncotarget.4228. PMC 4673253. PMID 26053184.
  237. Gastl G, Plante M (2001). "Bioactive interleukin-6 levels in serum and ascites as a prognostic factor in patients with epithelial ovarian cancer". Methods Mol. Med. 39: 121–3. doi:10.1385/1-59259-071-3:121. PMID 21340762.
  238. 238.0 238.1 Colvin EK (2014). "Tumor-associated macrophages contribute to tumor progression in ovarian cancer". Front Oncol. 4: 137. doi:10.3389/fonc.2014.00137. PMC 4047518. PMID 24936477.
  239. Mendonça FM, de Sousa FR, Barbosa AL, Martins SC, Araújo RL, Soares R, Abreu C (February 2015). "Metabolic syndrome and risk of cancer: which link?". Metab. Clin. Exp. 64 (2): 182–9. doi:10.1016/j.metabol.2014.10.008. PMID 25456095.
  240. Diaz ES, Karlan BY, Li AJ (May 2013). "Obesity-associated adipokines correlate with survival in epithelial ovarian cancer". Gynecol. Oncol. 129 (2): 353–7. doi:10.1016/j.ygyno.2013.02.006. PMID 23402904.
  241. Coosemans A, Baert T, Vergote I (2015). "A view on dendritic cell immunotherapy in ovarian cancer: how far have we come?". Facts Views Vis Obgyn. 7 (1): 73–8. PMC 4402447. PMID 25897374.
  242. Coosemans A, Vergote I, Van Gool SW (December 2013). "Dendritic cell-based immunotherapy in ovarian cancer". Oncoimmunology. 2 (12): e27059. doi:10.4161/onci.27059. PMC 3913669. PMID 24501688.
  243. Morehead LC, Cannon MJ (August 2018). "Further clinical advancement of dendritic cell vaccination against ovarian cancer". Ann Res Hosp. 2. doi:10.21037/arh.2018.08.02. PMC 6192055. PMID 30345421.
  244. Curiel TJ, Coukos G, Zou L, Alvarez X, Cheng P, Mottram P, Evdemon-Hogan M, Conejo-Garcia JR, Zhang L, Burow M, Zhu Y, Wei S, Kryczek I, Daniel B, Gordon A, Myers L, Lackner A, Disis ML, Knutson KL, Chen L, Zou W (September 2004). "Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival". Nat. Med. 10 (9): 942–9. doi:10.1038/nm1093. PMID 15322536.
  245. Schmidt FM, Weschenfelder J, Sander C, Minkwitz J, Thormann J, Chittka T, Mergl R, Kirkby KC, Faßhauer M, Stumvoll M, Holdt LM, Teupser D, Hegerl U, Himmerich H (2015). "Inflammatory cytokines in general and central obesity and modulating effects of physical activity". PLoS ONE. 10 (3): e0121971. doi:10.1371/journal.pone.0121971. PMC 4363366. PMID 25781614.
  246. Zhang M, He Y, Sun X, Li Q, Wang W, Zhao A, Di W (February 2014). "A high M1/M2 ratio of tumor-associated macrophages is associated with extended survival in ovarian cancer patients". J Ovarian Res. 7: 19. doi:10.1186/1757-2215-7-19. PMC 3939626. PMID 24507759.
  247. 247.0 247.1 247.2 Liu Y, Metzinger MN, Lewellen KA, Cripps SN, Carey KD, Harper EI, Shi Z, Tarwater L, Grisoli A, Lee E, Slusarz A, Yang J, Loughran EA, Conley K, Johnson JJ, Klymenko Y, Bruney L, Liang Z, Dovichi NJ, Cheatham B, Leevy WM, Stack MS (December 2015). "Obesity Contributes to Ovarian Cancer Metastatic Success through Increased Lipogenesis, Enhanced Vascularity, and Decreased Infiltration of M1 Macrophages". Cancer Res. 75 (23): 5046–57. doi:10.1158/0008-5472.CAN-15-0706. PMC 4668203. PMID 26573796.
  248. 248.0 248.1 Preston CC, Goode EL, Hartmann LC, Kalli KR, Knutson KL (April 2011). "Immunity and immune suppression in human ovarian cancer". Immunotherapy. 3 (4): 539–56. doi:10.2217/imt.11.20. PMC 3147144. PMID 21463194.
  249. Naylor C, Petri WA (February 2016). "Leptin Regulation of Immune Responses". Trends Mol Med. 22 (2): 88–98. doi:10.1016/j.molmed.2015.12.001. PMID 26776093.
  250. Yang C, Lee H, Jove V, Deng J, Zhang W, Liu X, Forman S, Dellinger TH, Wakabayashi M, Yu H, Pal S (2013). "Prognostic significance of B-cells and pSTAT3 in patients with ovarian cancer". PLoS ONE. 8 (1): e54029. doi:10.1371/journal.pone.0054029. PMC 3542323. PMID 23326565.
  251. Dong HP, Elstrand MB, Holth A, Silins I, Berner A, Trope CG, Davidson B, Risberg B (March 2006). "NK- and B-cell infiltration correlates with worse outcome in metastatic ovarian carcinoma". Am. J. Clin. Pathol. 125 (3): 451–8. PMID 16613351.
  252. Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G (February 2012). "Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis". Gynecol. Oncol. 124 (2): 192–8. doi:10.1016/j.ygyno.2011.09.039. PMC 3298445. PMID 22040834.
  253. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, Lele S, Chen YT, Ohtani H, Old LJ, Odunsi K (December 2005). "Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer". Proc. Natl. Acad. Sci. U.S.A. 102 (51): 18538–43. doi:10.1073/pnas.0509182102. PMC 1311741. PMID 16344461.
  254. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G (January 2003). "Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer". N. Engl. J. Med. 348 (3): 203–13. doi:10.1056/NEJMoa020177. PMID 12529460.
  255. Hansen JM, Coleman RL, Sood AK (March 2016). "Targeting the tumour microenvironment in ovarian cancer". Eur. J. Cancer. 56: 131–143. doi:10.1016/j.ejca.2015.12.016. PMC 4769921. PMID 26849037.
  256. Wang B, Wood IS, Trayhurn P (December 2007). "Dysregulation of the expression and secretion of inflammation-related adipokines by hypoxia in human adipocytes". Pflugers Arch. 455 (3): 479–92. doi:10.1007/s00424-007-0301-8. PMC 2040175. PMID 17609976.
  257. Slaughter KN, Thai T, Penaroza S, Benbrook DM, Thavathiru E, Ding K, Nelson T, McMeekin DS, Moore KN (April 2014). "Measurements of adiposity as clinical biomarkers for first-line bevacizumab-based chemotherapy in epithelial ovarian cancer". Gynecol. Oncol. 133 (1): 11–5. doi:10.1016/j.ygyno.2014.01.031.
  258. Spentzos D, Cannistra SA, Grall F, Levine DA, Pillay K, Libermann TA, Mantzoros CS (September 2007). "IGF axis gene expression patterns are prognostic of survival in epithelial ovarian cancer". Endocr. Relat. Cancer. 14 (3): 781–90. doi:10.1677/ERC-06-0073. PMID 17914107.
  259. Pollak MN, Schernhammer ES, Hankinson SE (July 2004). "Insulin-like growth factors and neoplasia". Nat. Rev. Cancer. 4 (7): 505–18. doi:10.1038/nrc1387. PMID 15229476.
  260. Laws MJ, Kannan A, Pawar S, Haschek WM, Bagchi MK, Bagchi IC (March 2014). "Dysregulated estrogen receptor signaling in the hypothalamic-pituitary-ovarian axis leads to ovarian epithelial tumorigenesis in mice". PLoS Genet. 10 (3): e1004230. doi:10.1371/journal.pgen.1004230. PMC 3945209. PMID 24603706.
  261. Schairer C, Fuhrman BJ, Boyd-Morin J, Genkinger JM, Gail MH, Hoover RN, Ziegler RG (January 2016). "Quantifying the Role of Circulating Unconjugated Estradiol in Mediating the Body Mass Index-Breast Cancer Association". Cancer Epidemiol. Biomarkers Prev. 25 (1): 105–13. doi:10.1158/1055-9965.EPI-15-0687. PMC 5555590. PMID 26637268.