Next Article in Journal
Physical, Chemical and Biochemical Modifications of Protein-Based Films and Coatings: An Extensive Review
Next Article in Special Issue
NR2F6 Expression Correlates with Pelvic Lymph Node Metastasis and Poor Prognosis in Early-Stage Cervical Cancer
Previous Article in Journal
Heads or Tails: Genotyping of Hepatitis C Virus Concerning the 2k/1b Circulating Recombinant Form
Previous Article in Special Issue
Growth Hormone Releasing Peptide-2 Attenuation of Protein Kinase C-Induced Inflammation in Human Ovarian Granulosa Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells

Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42, Milan 20133, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(9), 1387; https://doi.org/10.3390/ijms17091387
Submission received: 14 July 2016 / Revised: 11 August 2016 / Accepted: 13 August 2016 / Published: 23 August 2016
(This article belongs to the Special Issue Gynecologic Oncology: From Molecular Mechanisms to Targeted Therapies)

Abstract

:
Epithelial ovarian cancer (EOC) is the deadliest tumor among gynecological cancer in the industrialized countries. The EOC incidence and mortality have remained unchanged over the last 30 years, despite the progress in diagnosis and treatment. In order to develop novel and more effective therapeutic approaches, the molecular mechanisms involved in EOC progression have been thoroughly investigated in the last few decades. At the late stage, peritoneal metastases originate from the attachment of small clusters of cancer cells that shed from the primary site and carried by the ascites adhere to the abdominal peritoneum or omentum. This behavior suggests that cell–cell or cell–matrix adhesion mechanisms regulate EOC growth and dissemination. Complex downstream signalings, which might be influenced by functional cross-talk between adhesion molecules and co-expressed and activated signaling proteins, can affect the proliferation/survival and the migration/invasion of EOC cells. This review aimed to define the impact of the mechanisms of cell–cell, through cadherins, and cell–extracellular matrix adhesion, through integrins, on the signaling cascades induced by membrane receptors and cytoplasmic proteins known to have a role in the proliferation, migration and invasion of EOC cells. Finally, some novel approaches using peptidomimetic ligands to cadherin and integrins are summarized.

Graphical Abstract

1. Introduction

Epithelial ovarian cancer (EOC) is a devastating disease with an overall five-year survival rate of approximately 45% [1,2]. EOCs are usually diagnosed when malignant cells have already invaded the peritoneal cavity and, although most of the patients are sensitive to the first line chemotherapy, 50% of them relapse with a chemoresistant disease. For all these reasons, EOCs are the fifth main cause of cancer-related deaths among women, and the primary cause of death from gynecological cancer [3]. Therefore, in cancer research, investigations aiming to clarify the mechanisms of EOC tumorigenesis and progression are one of the most important areas. EOC group different diseases with a common anatomical location [4] but display high molecular and etiological heterogeneity [5,6,7,8].
EOCs are divided into two large groups [9] designated types I, genetically stable and not very aggressive [10], and type II, genetically unstable and very aggressive tumors, which are usually diagnosed at the advanced-stage. Type II tumors include high-grade serous, high-grade endometrioid, malignant mixed mesodermal tumors (carcinosarcomas), and undifferentiated carcinomas being the serous high-grade ovarian carcinoma (HGSOC) the most representative tumors [11].
More than 50% of EOC patients at the late stage, in particular those with type-II tumors, present with ascites/effusions in their abdominal cavity rich of tumor cells [12]. The ascites are accumulated since implanted tumor cells give rise to the obstruction of lymphatic vessels, preventing the outflow of fluid that transpires from the tumor vessels. Hence, patients affected by HGSOC type-II tumors have the peritoneal cavity invaded by metastatic tumors, growing in the solid stromal matrices, and multicellular aggregates (MCAs) floating and growing in the malignant ascites [13]. These MCAs overcome anoikis [14] and persist as ascites [15]. Although a possible mechanism of hematogenous HGSOC metastasis formation to the omentum has also been reported [16], the general consensus is that these MCAs originate by the shedding of malignant cells into the peritoneum from the primary tumor and that disaggregation and attachment to the sub-mesothelial extracellular matrix (ECM) allow the formation of secondary lesions [17,18,19]. Once adhered to the peritoneum, EOC cells proliferate, migrate and invade the surrounding tissues. An elegant approach of a live image-based in vitro model determined that a myosin–generated force allows EOC MCAs to displace and remove the mesothelial monolayer. This process is now known as mesothelial cell clearance [20]. The EOC metastasis outgrowth occurs upon the remodeling of cell–cell adhesion molecules (i.e., cadherins) during spheroids dis-aggregation. In addition, the integrins expressed on the surface of EOC cells are essential to the attachment of EOC cells to the sub-mesothelial ECM [18,19,20,21].
Interestingly, a proteomic approach for MCA analysis for EOC patient stratification has been able to identify three adhesion-related subsets with potential predictive impact [22]. These data highlight the influence of the adhesion molecules in the clinical EOC behavior.
Overall, the processes of EOC progression require that cell–cell, through cadherins, and cell–ECM adhesion, through integrins, cooperate, directly or indirectly, to the activation of signaling pathways relevant to the proliferation/survival and the migration/invasion mechanisms of EOC cells.

2. Cadherin-Associated Signaling Activation

Cell–cell adhesion is mediated by cadherins (cadhs) through the calcium-dependent homophilic interaction of their extracellular domain to form, associated with cytoplasmic proteins (β- and α-catenin (ctn) together with p120ctn) intercellular structures called adherens junctions (AJs) (see, for review, [23]). The AJs constitute a physical bridge between the cadherin complex and the cortical actin filaments thus regulating cellular dynamic behaviors, such as rearrangements, movement and shape changes during embryonic development as well as during neoplastic transformation and progression. The cytoplasmic tail of cadhs has no catalytic activity; therefore, possible signaling activation must occur upon the recruitment of signaling molecules to the site of the cadh–ctn complex [24]. In epithelial tissues, the homophilic E-cadh ligation can activate the junctional GTPase signaling downstream to Rac [25,26,27,28] and Cdc42 [26,29]. E-cadh can therefore regulate the localization and function of the Rho GTPase with a mechanism described as ‘outside-in signaling’ [30,31] (Figure 1a).
Below, E- and P-cadh-associated signalings are discussed. N-cadh displays similar structural and functional properties to E-cadh (see, for review, [32]). In EOCs, N-cadh expression is associated with a more aggressive and chemoresistant phenotype [33], but its role in modulating signaling activation is still unclear.

2.1. E-cadh

In cancer, the switch in expression of E-cadh to N-cadh or the up-regulation of cadh-11 and P-cadh are important indicators of progression and are believed to facilitate the epithelial–mesenchymal transition (EMT), which leads to a more migratory and invasive phenotype [34,35]. In metastatic solid tumors, E-cadh expression is usually lost in human breast, bladder, lung and pancreatic carcinomas, which nevertheless express other cadherins, especially N-cadh [36]. For this reason, E-cadh has been proposed as a tumor suppressor since, in invasive carcinoma cells, ectopically expressed E-cadh was associated with a decreased growth potential [37]. In confluent cells, E-cadh was shown to be associated to epidermal growth factor receptor (EGFR) leading to receptor immobilization and alteration of the ligand-receptor affinity, thus inhibiting the signaling activation [38,39].
Very recent publications are beginning to revisit the general consensus that EMT is necessary for the metastasis formation occurring when the tumors become more malignant. In these reports, E-cadh expression was found maintained during the formation of lung metastasis in mouse models of breast and pancreatic cancers [40,41]. These data are not surprising since E-cadh lost was previously showed to be a rare event in both invasive ductal [42] and inflammatory breast carcinomas [43]. Mechanistically, in a breast carcinoma patient-derived 3D organoid model, E-cadh homophilic adhesion was present in a leading cell population and necessary during invasion of collagen I gel [44,45].
In EOCs, initial immunohistochemical analysis revealed low E-cadh expression in advanced-stage disease [46,47]. Data made on a higher number of EOC biopsies assessed that E-cadh is still expressed at cell–cell contacts in late stage tumors [48,49,50,51]. Furthermore, in EOC MCAs, the predominant cadherin expressed at cell–cell contact is E-cadh (our unpublished results and [52]) and contributes to aggregate formation [53]. E-cadh was first assessed to upregulate ligand-independent EGFR trans-phosphorylation leading to AKT and MAPK activations [54] (Figure 1a). Furthermore, E-cadh-mediated AJ formation recruits PI3K-p85 to the cell membrane contributing to activation of the PI3K/AKT pathway and to the up-modulation of EOC growth [55]. In the EOC cell line SKOV3, E-cadh functionality can also increase mitogen-activated protein kinase/extracellular-signal regulated kinase (MEK/ERK) activation, although no physical association with MEK or even EGFR was demonstrated [56] (Figure 1b).
While in normal epithelial cells E-cadh is maintained on the membrane by p120 catenin and its levels are controlled through the endocytic pathway [57], in transformed cells, E-cadh loss is also due to the proteolitic cleavage of the extra-cellular domain resulting in a 80-KDa fragment. This soluble (s) E-cadh might be generated though the action of metalloproteases (MMTs), such as ADAM10 and MMPs [58,59] usually upregulated in cancers, or γ-secretases, like presenilin 1 [60]. The 80 KDa soluble form of E-cadh has been found in the serum of cancer patients with poor prognosis [61]. In EOCs, the levels of sE-cadh in cystic fluid from patients presenting with complex ovarian masses was higher than in patients presenting with benign tumors [60]. sE-cadh was found in the EOC ascites, and in vitro experiments demonstrated that the integrin clustering occurring during the adhesion to the sub-mesothelial collagen I stimulated EGFR-dependent MMP9 expression [62], thus inducing sE-cadh production [63]. These data suggest that EGFR activation and production of sE-cadh constitute a feedback loop able to promote proliferation and/or migration/invasion. More recently, the 85-KDa and 23-KDa E-cadh, produced by calpain, were found in solid metastatic peritoneal masses of advanced EOC patients [64]. The presence of the 85-KDa E-cadh fragment correlated with a worse patient outcome, and the authors hypothesized that the 85-KDa fragment might enable tumor cluster formation and peritoneal dissemination [64].

2.2. P-cadh

Among cadherins, P-cadh has a proven role during the progression of several carcinomas, as cervix [65], esophagous [66], breast [67] and colon [68], including EOCs [69]. In EOC, an increase in P-cadh expression in the tumor masses, concomitant with a decrease of E-cadh expression, is associated with a progression from stage I to stage II tumors [69]. The pro-metastatic role of P-cadh is mainly due to a cross-talk with the axis constituted by the cytoplasmic p120ctn and the activation of the Rho GTPase signaling. In this context, once Gonadotropin-releasing hormone (GnRH), whose physiologic role is the control of pituitary gonadotropin secretion, induces the E- to P-cadh switching, p120ctn moves from the membrane to the cytoplasm, thus leading to activation of mechanisms of migration and invasion [70]. The role of p120ctn as determinant of migration upon GnRH stimulation is due to a trans-activation of insulin-like growth factor-1 receptor (IGF-1R) by P-cadh, which induces Rac1 and Cdc42 activations [71].
P-cadh also contributes to the adhesion of EOC cells to the peritoneum and in the metastasis formation and has been considered the predominant cadherin subtype expressed in the MCAs of the peritoneal effusion [69]. Furthermore, its inhibition reduced tumor growth, ascites formation and metastatis in in vivo pre-clinical models [72]. Besides its role in the MCA formation, P-cadh can also contribute to inhibition of anoikis. Accordingly, in vitro P-cadh knockdown increases cell death of EOC cells growth in suspension and in vivo decreases the dissemination of EOC cells injected in the peritoneum of immunocompromised mice [73], once again through a cross-talk with p120ctn and Rho GTPase activation. Another report has recently showed a P-cadh role in the MCA formation through a cross-talk with β1 integrin. In particular, P-cadh induces the Golgi glycosyltransferase (ST6Gal-1), which mediate β1 integrin hypersialylation, a post-translational modification that results in maturation of this integrin and activation of the downstream signaling [74] (Figure 1b).

3. Integrin-Associated Signaling Activation

Integrins are αβ heterodimeric transmembrane proteins implicated in numerous physiological processes including adhesion to the extracellular matrix, proliferation, survival, migration and differentiation [75]. The molecular and physical interactions among the cells and with ECM can strongly affect cell behavior, thus inducing cancer cell invasion and dissemination. A group of integrins recognizes and binds to the Arg-Gly-Asp (RGD) motif present on the ECM proteins, and the specificity of integrin binding to different ECM proteins is determined, in part, by other amino acids surrounding the RGD sequence [76]. As already said for the cadherins, ntegrin cytoplasmic tails do not exert a kinase activity but instead are able to activate specific intracellular kinases, such as FAK (Focal Adhesion Kinase), which, in turn, recruit the src kinase [77]. Src phosphorylates a number of FAK-associated proteins including paxilin, tensin and the adaptor p130CAS (Crk-Associated Substrate). FAK activation also leads to the recruitment of other SH2-containing proteins, including the PI3K, PLC-γ and the adapter proteins Grb7 [77] and Grb2 to partially mediate ERK activation [78]. Finally, the FAK/Src complex modulates the activity of small GTPases leading to the actin cytoskeleton remodeling necessary for cell adhesion and migration [79].
In EOC, the expression of ECM components and integrins has been found to be involved not only in the cell detachment from the primary tumor and from the peritoneal metastasis [80,81,82], but also in the resistance to anoikis of the EOC spheroids [83,84]. Dysregulation and aberrant deposition of ECM components, due to the activation of different MMPs [85], contribute to tumor progression. The expression of vitronectin [80] and fibronectin [86] at the periphery of mesothelial cells actively participates in EOC cell motility and metastasis. In addition, fibronectin has been also shown to mediate EOC cell migration and invasion through the upregulation of the FAK/PI3K/Akt pathway [87] and is an indicator of poor prognosis in invasive EOC [88].

3.1. Integrins and Receptor Tyrosine Kinase (RTKs)

Numerous studies suggest that the cooperation between integrins and RTKs exists and exerts a central role in cancer progression regulating invasion, proliferation and survival (see, for review, [89]). Considering that neither α or β subunit possess catalytic activity, it is possible that multiple mechanisms may regulate the cross-talk between integrins and RTKs. Three main types of integrins/RTKs interaction have been identified [90] (Figure 2): (1) integrins can physically bind to RTKs; (2) integrins clustering upon the binding to ECM can enhance signaling pathways that are activated following ligand-dependent RTKs activation; and (3) integrins and RTKs reciprocally control their surface expression.
The RTK represents the largest family of oncogenes, and the inhibitors targeting these receptors have already shown clinical efficacy as anti-cancer agents in solid tumors other than EOCs [91]. RTKs have been found to be over-expressed in EOCs as compared to normal counterparts [92].

3.1.1. EGFR

The EGFR is a transmembrane glycoprotein receptor that belongs to the ErbB family of RTKs. The ligands for the EGFR are the EGF, the betacellulin (BTC), TGF-α, amphiregulin (AR), and epiregulin (EPR) [93]. EGF ligands can induce the homodimerization of EGFR as well as the heterodimerization with other members of the family: HER2, HER3 and HER4 [93].
In solid tumors, EGFR can interact with many integrins such as β1 [94], α6β4 [95] and αvβ3 [96,97], likely by forming a multimeric complex that also includes Src and the adaptor protein p130Cas [96]. This complex is necessary for the ligand-independent activation of the EGFR leading to signaling involved in cell survival and proliferation in response to ECM [94]. EGFR is expressed in up to 70% of EOCs [98], and its activation contributes to cell proliferation, invasion, angiogenesis and resistance to apoptosis [99]. EGFR activation can also induce a pro-inflammatory program with the co-expression of IL-6 and PAI-1 in a subset of EGFR-expressing EOCs with shorter progression-free survival after chemotherapy [100]. A direct correlation between EGFR and integrin expression has been reported. Indeed, reduced expression of EGFR in NIH:OVCAR-8 cells was associated with reduced integrin α6, a laminin-1 receptor, and cell adhesion with the down-modulation of MMP-9 activity, thus reducing the aggressiveness of EOC cells [101]. Conversely, the expression of a constitutively active EGFR has been proposed to contribute to a more aggressive disease throughout the down-modulation of the integrin α2 expression, which leads to changes in cell shape and focal adhesion formation [102].

3.1.2. c-MET

c-MET oncogene is a membrane receptor that is essential for embryonic development and wound healing [103]. The natural ligand for this receptor is the hepatocyte growth factor, HGF, whose binding to the c-MET receptor leads to receptor phosphorylation and activation [103].
As in other carcinomas, also in EOCs, Sawada et al. found that the over-expression of c-MET was associated with the worst prognosis [104]. The knockdown of c-Met expression by small interfering RNA (siRNA) induced a decreased activation of the MEK/ERK and PI3K/AKT signaling pathways and reduced adhesion, invasion, peritoneal dissemination, and tumor growth through the inhibition of the expression of α5β1 integrin [104]. Furthermore, α5β1 integrin binding to the fibronectin also leads to a ligand-independent activation of c-MET signaling to FAK and Src. This mechanism is due to a biochemical association between α5 integrin and c-MET [105]. Since it has been reported that cancer-associated mesothelial cells that colonize the peritoneum produce fibronectin [86], able to trap HGF produced by the fibroblasts of the tumor microenvironment [106], the inhibition of c-MET activation seems to be a promising target to block cancer metastasis. Recently, Moran-Jones et al. investigated the effect of a novel ATP competitive inhibitor, INC280 (Novartis, Basel, Switzerland), which selectively inhibits c-MET activation in a number of EOC in vitro models [107], also decreasing the migration of EOC cells adherent to ex vivo peritoneal tissue.

3.1.3. VEGFR Family

For EOC patients, targeting angiogenesis is a novel therapeutic option, which includes the use of bevacizumab, an antibody that binds to VEGF, thus inhibiting its association with the receptor [108]. The VEGF and the VEGFR are expressed by EOC cells, and increased VEGF expression has been associated with the tumor progression and poor survival [109,110].
The interaction between the VEGFR and integrins is very important for angiogenesis. In particular, ligand-dependent VEGFR-2 phosphorylation activates c-src which then phosphorylates the cytosolic tail of β3 integrin, promoting a feedback loop with the formation of the VEGFR-2/ανβ3 complex and increased integrin and VEGFR-2 signaling [111].
In in vivo EOC models, the combination of anti-angiogenic and anti-integrin combination therapy using Bevacizumab and etaracizumab (an antibody that binds to the integrin αvβ3 inhibiting its activation) was more effective in inhibiting tumor growth and microvessel density than individual drugs [112]. In particular, the etaracizumab was reported to be particularly efficient in reducing the volumes of VEGFR-dependent SKOV3ip tumors in nude mice [113]. These findings might open the possibility that a dual blockade of VEGF and the αVβ3 integrin in combination with chemotherapy may impair EOC growth in those patients whose tumors display high vessel density.

3.1.4. Axl

Deregulation of the receptor tyrosine kinase Axl, a member of the TAM RTK family, which also includes Mer and Tyro3, has been implicated in aggressive phenotype, tumor metastases and the progression of several human cancers [114]. The ligand of TAM RTKs, Gas6, displays the highest activity for Axl, which has been proposed as a therapeutic target since its genetic and pharmacologic inhibition could prevent or overcome acquired resistance to EGFR inhibitors [115]. Indeed, an EMT signature including Axl was identified and associated with the resistance to EGFR and PI3K inhibitors [116]. Multiple downstream signaling pathways, including PI3K/Akt and MAPK, can be elicited by Axl/Gas6 in different cell types [117,118].
Axl expression increased in type II EOC primary tumors and metastases compared to the ovarian surface epithelium [119] and Gas6 has been identified as prognostic marker in HGEOCs [120]. Therapeutic blockade of Axl activation reduced the migratory and invasive capability of HGSOC cells in vitro and in xenograft models [121]. Recently, the signaling elicited by Axl activation was dissected showing a cross-talk with the integrin/ECM pathway through the adaptor protein p130Cas leading to Rac activation [122]. Furthermore, an Axl-driven 61-gene signature, which included several collagens and ECM-associated proteins, was associated to the most aggressive HGSOC subtype [122].

3.2. Integrins and RNAseT2

A novel factor that may play a role in the complex integrin signaling is the protein RNASET2, a ribonuclease that belongs to the T2 family. This protein, known to be downregulated in EOC and EOC cell lines [123], has been found to display tumorigenic and metastatic suppressor properties, independent from its ribonuclease activity, in vivo [123,124,125,126]. Recently, a new role of this protein was discovered that led to the hypothesis than RNASET2 may affect the integrin signaling pathway. RNASET2 proved to have the capability to reorganize the actin cytoskeleton and consequently influence cells motility, invasion capability and cell adhesion to ECM. In fact, silencing of the protein in the EOC cell line NIH-OVCAR3 triggered a disruption of the network of actin filaments and stress fibers, inducing a pattern of peripheral actin filaments [126], thus leading to increased migration and adhesion to laminin, collagen I and IV. Of note, the adhesion of these cells on collagen I was accompanied to an increase of paxillin activation and, accordingly, to an increase of mature focal adhesions (FAs) [126]. Although RNASET2 and tr-T2-50, a recombinant truncated form of the human RNASET2 deprived of its RNAse activity [127,128], showed the capability to bind actin in vitro, Acquati et al. did not find any co-localization between the actin cytoskeleton and RNASET2 in NIH-OVCAR3 cells [126], supporting the hypothesis of a cross-talk with the integrin activation and consequent re-organization of the actin cytoskeleton. Conversely, RNASET2 resulted in being significantly down-regulated in drug-resistant EOC biopsies and EOC cell lines, suggesting an involvement of this protein in the response to chemotherapy [129]. Since integrin signaling pathway is also associated with therapy resistance in cancer (see, for reviews, [130,131]) it can be assumed that RNASET2 may act on integrin signaling pathway, not only influencing FA dynamics, cell motility and adhesion on ECM, but also influencing downstream PI3K/AKT pathwaya and, in turn, drug resistance.

4. Targeting Cell Adhesion Using Peptidomimetic Ligands

Besides the use of specific antibodies, in the last few years, the possibility of designing small molecules acting as antagonists or agonists of the interactions between cell/ECM or cell/cell and the consequent intracellular signalings has been developed. Peptidomimetic integrin ligands have proved to be valuable for tumor directed delivery of diagnostics or therapeutics [132,133,134,135]. A subset of integrins (8 out of 24) recognizes RGD sequence and several RGD peptide mimetics entered in clinical trials as integrin-targeted agents in EOC [136]. Integrin ligands tested in tumor cells are summarized in Table 1. The cilengitide, designed against the αvβ3, has been considered the most promising compound; however, the phase III clinical trials failed likely due to a low specificity, metabolic instability and low biodistribution [137,138]. To accomplish an adequate biodistribution and uptake in cancer cells, new compounds were obtained by the coniugation between the peptidomemitic ligands and a drug or a nanoparticle made of new materials (see, for review, [139]).
As far as the cadherins are concerned, some short peptide sequences, corresponding to the binding domain of N-cadh, have been shown to disrupt cell adhesion-induced apoptosis in an antagonist manner [140] (Table 1). In cancer cells in vitro and in xenotransplanted animal models, one of these small peptides, the ADH-1, enhanced antitumor activity of melphalan-treated melanomas [141] by altering both homotypic (between cancer cells) and heterotypic (between cancer cells and surrounding endothelial cells) cadherin interactions. Although ADH-1 has been well tolerated in phase I and II clinical trials, its antitumor efficacy as a single agent has been moderate [142,143]. This is probably due to its low affinity for N-cadh and the lack of stability in biological fluids. Very recently, Turley et al. assessed that ADH-1 may have a dichotomous effect: it can increase tumor growth rate and sensitivity to some chemotherapy agents by increasing AKT activation and, on the other side, ADH-1 facilitates drug delivery, enhancing the vascular permeability [144]. More recently, some peptidomimetics able to bind at the sites of the homophilic N-and E-cadh binding have been shown to inhibit cell–cell adhesion [145] and E-cadherin-mediated AKT phosphorylation of EOC cells [146].

5. Conclusions

The present review has reported the more recent knowledge on the cooperation of cell–cell and cell–ECM adhesion molecules, such as cadherins and integrins, as well as their elicited mechanisms, together with membrane or cytoplasmic signaling molecules that are required for the growth, migration and invasion of EOC cells, especially taking into account those mechanisms necessary for EOC intra-peritoneal dissemination. The reported information suggests that different cadherins and integrins could affect several aspects of EOC progression. For example, E-cadh is important in spheroid formation, but, at the late stage, the switch in cadherins expression could enhance metastasis through a different pattern of integrins at the plasma membrane.
Recent evidence shows that mono-targeting, particularly of RTKs, inefficiently impacts tumor growth control, suggesting that a multi-target treatment might be more efficient. As we have shown here, RTKs might require specific integrins for full activation, and for their contribution to tumorigenesis and metastasis formation, it may be plausible to tailor the use of integrin antagonists in combination with RTK inhibitors. For instance, inhibitors to adhesion molecules together with target-specific drugs could be effective as adjuvant therapy in advanced-stage EOC patients. Peptidomimetics to E-cadh could be relevant for EOC relapsing patients presenting E-cadh-expressing MCAs, and labelled E-cadh binding compound/s could be exploited for the detection of minimal residual disease after first debulking or during chemotherapeutic treatment. Overall, these new approaches, aimed to inhibit adhesion-dependent mechanisms, represent a challenge in the field of cancer research likely to counteract the processes of mesothelial cell clearance and MCA formation occurring during the intra-peritoneal dissemination of EOC cells. Investigations through large scale/high-throughput technologies could help to identify further relevant cross-talks that involve adhesion molecules.
Further efforts are necessary for a better understanding of the different roles exerted by the numerous players participating in these complex mechanisms for a more rational development of better treatments in the aim to improve EOC patient outcome.

Acknowledgments

The authors acknowledge Anna Maria Invernizzi for her collaboration in the development of the projects in the EOC field. This work was supported by the Associazione Italiana per La Ricerca sul Cancro (AIRC, IG13055 to Antonella Tomassetti and IG17475 to Delia Mezzanzanica), by the Italian Ministry of Health (′Characterization of the molecular basis of tumor progression: a focus on ovarian cancer′ Ricerca Corrente 2013–2016 to Antonella Tomassetti) and the Cariplo Foundation (2013-0865 to Delia Mezzanzanica).

Author Contributions

All authors contributed equally to the preparation of the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jelovac, D.; Armstrong, D.K. Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J. Clin. 2011, 61, 183–203. [Google Scholar] [CrossRef] [PubMed]
  2. Baldwin, L.A.; Huang, B.; Miller, R.W.; Tucker, T.; Goodrich, S.T.; Podzielinski, I.; DeSimone, C.P.; Ueland, F.R.; van Nagell, J.R.; Seamon, L.G. Ten-year relative survival for epithelial ovarian cancer. Obstet. Gynecol. 2012, 120, 612–618. [Google Scholar] [CrossRef] [PubMed]
  3. Jayson, G.C.; Kohn, E.C.; Kitchener, H.C.; Ledermann, J.A. Ovarian cancer. Lancet 2014, 384, 1376–1388. [Google Scholar] [CrossRef]
  4. Vaughan, S.; Coward, J.I.; Bast, R.C., Jr.; Berchuck, A.; Berek, J.S.; Brenton, J.D.; Coukos, G.; Crum, C.C.; Drapkin, R.; Etemadmoghadam, D.; et al. Rethinking ovarian cancer: Recommendations for improving outcomes. Nat. Rev. Cancer 2011, 11, 719–725. [Google Scholar] [CrossRef] [PubMed]
  5. Tothill, R.W.; Tinker, A.V.; George, J.; Brown, R.; Fox, S.B.; Lade, S.; Johnon, D.S.; Trivett, M.K.; Etemadmoghadam, D.; Locandro, B.; et al. Novel molecular subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin. Cancer Res. 2008, 14, 5198–5208. [Google Scholar] [CrossRef] [PubMed]
  6. The Cancer Genome Atlas Database. 2012. Available online: https://tcga-data.nci.nih.gov/ (accessed on 30 June 2011).
  7. Sieh, W.; Salvador, S.; McGuire, V.; Weber, R.P.; Terry, K.L.; Rossing, M.A.; Risch, H.; Wu, A.H.; Webb, P.M.; Moysich, K.; et al. Tubal ligation and risk of ovarian cancer subtypes: A pooled analysis of case-control studies. Int. J. Epidemiol. 2013, 42, 579–589. [Google Scholar] [CrossRef] [PubMed]
  8. Munksgaard, P.S.; Blaakaer, J. The association between endometriosis and ovarian cancer: A review of histological, genetic and molecular alterations. Gynecol. Oncol. 2012, 124, 164–169. [Google Scholar] [CrossRef] [PubMed]
  9. Kurman, R.J.; Shih, I.M. Pathogenesis of ovarian cancer: Lessons from morphology and molecular biology and their clinical implications. Int. J. Gynecol. Pathol. 2008, 27, 151–160. [Google Scholar] [CrossRef] [PubMed]
  10. Zorn, K.K.; Bonome, T.; Gangi, L.; Chandramouli, G.V.; Awtrey, C.S.; Gardner, G.J.; Barrett, J.C.; Boyd, J.; Birrer, M.J. Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer. Clin. Cancer Res. 2005, 11, 6422–6430. [Google Scholar] [CrossRef] [PubMed]
  11. Bowtell, D.D.; Bohm, S.; Ahmed, A.A.; Aspuria, P.J.; Bast, R.C., Jr.; Beral, V.; Berek, J.S.; Birrer, M.J.; Blagden, S.; Bookman, M.A.; et al. Rethinking ovarian cancer II: Reducing mortality from high-grade serous ovarian cancer. Nat. Rev. Cancer 2015, 15, 668–679. [Google Scholar] [CrossRef] [PubMed]
  12. Kipps, E.; Tan, D.S.; Kaye, S.B. Meeting the challenge of ascites in ovarian cancer: New avenues for therapy and research. Nat. Rev. Cancer 2013, 13, 273–282. [Google Scholar] [CrossRef] [PubMed]
  13. Ahmed, N.; Stenvers, K.L. Getting to know ovarian cancer ascites: Opportunities for targeted therapy-based translational research. Front. Oncol. 2013, 3, 256. [Google Scholar] [CrossRef] [PubMed]
  14. Tan, D.S.; Agarwal, R.; Kaye, S.B. Mechanisms of transcoelomic metastasis in ovarian cancer. Lancet Oncol. 2006, 7, 925–934. [Google Scholar] [CrossRef]
  15. Freedman, R.S.; Deavers, M.; Liu, J.; Wang, E. Peritoneal inflammation—A microenvironment for Epithelial Ovarian Cancer (EOC). J. Transl. Med. 2004, 2, 23. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Pradeep, S.; Kim, S.W.; Wu, S.Y.; Nishimura, M.; Chaluvally-Raghavan, P.; Miyake, T.; Pecot, C.V.; Kim, S.J.; Choi, H.J.; Bischoff, F.Z.; et al. Hematogenous metastasis of ovarian cancer: Rethinking mode of spread. Cancer Cell 2014, 26, 77–91. [Google Scholar] [CrossRef] [PubMed]
  17. Birbeck, M.S.; Wheatley, D.N. An electron microscopy study of the invasion of ascites tumor cells into the abdominal wall. Cancer Res. 1965, 25, 490–497. [Google Scholar] [PubMed]
  18. Shield, K.; Riley, C.; Quinn, M.A.; Rice, G.E.; Ackland, M.L.; Ahmed, N. α2β1 Integrin affects metastatic potential of ovarian carcinoma spheroids by supporting disaggregation and proteolysis. J. Carcinog. 2007, 6, 11. [Google Scholar] [CrossRef] [PubMed]
  19. Burleson, K.M.; Hansen, L.K.; Skubitz, A.P. Ovarian carcinoma spheroids disaggregate on type I collagen and invade live human mesothelial cell monolayers. Clin. Exp. Metastasis 2004, 21, 685–697. [Google Scholar] [CrossRef] [PubMed]
  20. Iwanicki, M.P.; Davidowitz, R.A.; Ng, M.R.; Besser, A.; Muranen, T.; Merritt, M.; Danuser, G.; Ince, T.A.; Brugge, J.S. Ovarian cancer spheroids use myosin-generated force to clear the mesothelium. Cancer Discov. 2011, 1, 144–157. [Google Scholar] [CrossRef] [PubMed]
  21. Barbolina, M.V.; Moss, N.M.; Westfall, S.D.; Liu, Y.; Burkhalter, R.J.; Marga, F.; Forgacs, G.; Hudson, L.G.; Stack, M.S. Microenvironmental regulation of ovarian cancer metastasis. Cancer Treat. Res. 2009, 149, 319–334. [Google Scholar] [PubMed]
  22. Kim, G.; Davidson, B.; Henning, R.; Wang, J.; Yu, M.; Annunziata, C.; Hetland, T.; Kohn, E.C. Adhesion molecule protein signature in ovarian cancer effusions is prognostic of patient outcome. Cancer 2012, 118, 1543–1553. [Google Scholar] [CrossRef] [PubMed]
  23. Takeichi, M. Dynamic contacts: Rearranging adherens junctions to drive epithelial remodelling. Nat. Rev. Mol. Cell Biol. 2014, 15, 397–410. [Google Scholar] [CrossRef] [PubMed]
  24. Shapiro, L.; Fannon, A.M.; Kwong, P.D.; Thompson, A.; Lehmann, M.S.; Grübel, G.; Legrand, J.-F.; Als-Nielsen, J.; Colman, D.R.; Hendrickson, W.A. Structural basis of cell-cell adhesion by cadherins. Nature 1995, 374, 327–337. [Google Scholar] [CrossRef] [PubMed]
  25. Kovacs, E.M.; Ali, R.G.; McCormack, A.J.; Yap, A.S. E-cadherin homophilic ligation directly signals through Rac and phosphatidylinositol 3-kinase to regulate adhesive contacts. J. Biol. Chem. 2002, 277, 6708–6718. [Google Scholar] [CrossRef] [PubMed]
  26. Kraemer, A.; Goodwin, M.; Verma, S.; Yap, A.S.; Ali, R.G. Rac is a dominant regulator of cadherin-directed actin assembly that is activated by adhesive ligation independently of Tiam1. Am. J. Physiol. Cell Physiol. 2007, 292, C1061–C1069. [Google Scholar] [CrossRef] [PubMed]
  27. Nakagawa, M.; Fukata, M.; Yamaga, M.; Itoh, N.; Kaibuchi, K. Recruitment and activation of Rac1 by the formation of E-cadherin-mediated cell-cell adhesion sites. J. Cell Sci. 2001, 114, 1829–1838. [Google Scholar] [PubMed]
  28. Yamada, S.; Nelson, W.J. Localized zones of Rho and Rac activities drive initiation and expansion of epithelial cell-cell adhesion. J. Cell Biol. 2007, 178, 517–527. [Google Scholar] [CrossRef] [PubMed]
  29. Kim, S.H.; Li, Z.; Sacks, D.B. E-cadherin-mediated cell-cell attachment activates Cdc42. J. Biol. Chem. 2000, 275, 36999–37005. [Google Scholar] [CrossRef] [PubMed]
  30. Fukata, M.; Kaibuchi, K. Rho-family GTPases in cadherin-mediated cell-cell adhesion. Nat. Rev. Mol. Cell Biol. 2001, 2, 887–897. [Google Scholar] [CrossRef] [PubMed]
  31. Wheelock, M.J.; Johnson, K.R. Cadherin-mediated cellular signaling. Curr. Opin. Cell Biol. 2003, 15, 509–514. [Google Scholar] [CrossRef]
  32. Oda, H.; Takeichi, M. Evolution: Structural and functional diversity of cadherin at the adherens junction. J. Cell Biol. 2011, 193, 1137–1146. [Google Scholar] [CrossRef] [PubMed]
  33. Miow, Q.H.; Tan, T.Z.; Ye, J.; Lau, J.A.; Yokomizo, T.; Thiery, J.P.; Mori, S. Epithelial-mesenchymal status renders differential responses to cisplatin in ovarian cancer. Oncogene 2014, 34, 1899–1907. [Google Scholar] [CrossRef] [PubMed]
  34. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
  35. Thiery, J.P.; Lim, C.T. Tumor dissemination: An EMT affair. Cancer Cell 2013, 23, 272–273. [Google Scholar] [CrossRef] [PubMed]
  36. Gheldof, A.; Berx, G. Cadherins and epithelial-to-mesenchymal transition. Prog. Mol. Biol. Transl. Sci. 2013, 116, 317–336. [Google Scholar] [PubMed]
  37. Wong, A.S.; Gumbiner, B.M. Adhesion-independent mechanism for suppression of tumor cell invasion by E-cadherin. J. Cell Biol. 2003, 161, 1191–1203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Qian, X.; Karpova, T.; Sheppard, A.M.; McNally, J.; Lowy, D.R. E-cadherin-mediated adhesion inhibits ligand-dependent activation of diverse receptor tyrosine kinases. EMBO J. 2004, 23, 1739–1748. [Google Scholar] [CrossRef] [PubMed]
  39. Andl, C.D.; Rustgi, A.K. No one-way street: Cross-talk between e-cadherin and receptor tyrosine kinase (RTK) signaling: A mechanism to regulate RTK activity. Cancer Biol. Ther. 2005, 4, 28–31. [Google Scholar] [CrossRef] [PubMed]
  40. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; El, R.T.; Ryu, S.; Troeger, J.; et al. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature 2015, 527, 472–476. [Google Scholar] [CrossRef] [PubMed]
  41. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature 2015, 527, 525–530. [Google Scholar] [CrossRef] [PubMed]
  42. Bertos, N.R.; Park, M. Breast cancer—One term, many entities? J. Clin. Investig. 2011, 121, 3789–3796. [Google Scholar] [CrossRef] [PubMed]
  43. Colpaert, C.G.; Vermeulen, P.B.; Benoy, I.; Soubry, A.; Van, R.F.; van, B.P.; Goovaerts, G.; Dirix, L.Y.; van, D.P.; Fox, S.B.; et al. Inflammatory breast cancer shows angiogenesis with high endothelial proliferation rate and strong E-cadherin expression. Br. J. Cancer 2003, 88, 718–725. [Google Scholar] [CrossRef] [PubMed]
  44. Cheung, K.J.; Gabrielson, E.; Werb, Z.; Ewald, A.J. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell 2013, 155, 1639–1651. [Google Scholar] [CrossRef] [PubMed]
  45. Nguyen-Ngoc, K.V.; Cheung, K.J.; Brenot, A.; Shamir, E.R.; Gray, R.S.; Hines, W.C.; Yaswen, P.; Werb, Z.; Ewald, A.J. ECM microenvironment regulates collective migration and local dissemination in normal and malignant mammary epithelium. Proc. Natl. Acad. Sci. USA 2012, 109, E2595–E2604. [Google Scholar] [CrossRef] [PubMed]
  46. Darai, E.; Scoazec, J.Y.; Walker-Combrouze, F.; Mlika-Cabanne, N.; Feldmann, G.; Madelenat, P.; Potet, F. Expression of cadherins in benign, borderline, and malignant ovarian epithelial tumors: A clinicopathologic study of 60 cases. Hum. Pathol. 1997, 28, 922–928. [Google Scholar] [CrossRef]
  47. Davies, B.R.; Worsley, S.D.; Ponder, B.A. Expression of E-cadherin, α-catenin and β-catenin in normal ovarian surface epithelium and epithelial ovarian cancers. Histopathology 1998, 32, 69–80. [Google Scholar] [CrossRef] [PubMed]
  48. Sundfeldt, K.; Piontkewitz, Y.; Ivarsson, K.; Nilsson, O.; Hellberg, P.; Brannstrom, M.; Janson, P.O.; Enerback, S.; Hedin, L. E-cadherin expression in human epithelial ovarian cancer and normal ovary. Int. J. Cancer 1997, 74, 275–280. [Google Scholar] [CrossRef]
  49. Imai, T.; Horiuchi, A.; Shiozawa, T.; Osada, R.; Kikuchi, N.; Ohira, S.; Oka, K.; Konishi, I. Elevated expression of E-cadherin and α-, β-, and γ-catenins in metastatic lesions compared with primary epithelial ovarian carcinomas. Hum. Pathol. 2004, 35, 1469–1476. [Google Scholar] [CrossRef] [PubMed]
  50. Hsia, D.A.; Mitra, S.K.; Hauck, C.R.; Streblow, D.N.; Nelson, J.A.; Ilic, D.; Huang, S.; Li, E.; Nemerow, G.R.; Leng, J.; et al. Differential regulation of cell motility and invasion by FAK. J. Cell Biol. 2003, 160, 753–767. [Google Scholar] [CrossRef] [PubMed]
  51. Tomassetti, A.; de Santis, G.; Castellano, G.; Miotti, S.; Mazzi, M.; Tomasoni, D.; van Roy, F.; Carcangiu, M.L.; Canevari, S. Variant HNF1 modulates epithelial plasticity of normal and transformed ovary cells. Neoplasia 2008, 10, 1481–1492. [Google Scholar] [CrossRef] [PubMed]
  52. Strauss, R.; Li, Z.Y.; Liu, Y.; Beyer, I.; Persson, J.; Sova, P.; Moller, T.; Pesonen, S.; Hemminki, A.; Hamerlik, P.; et al. Analysis of epithelial and mesenchymal markers in ovarian cancer reveals phenotypic heterogeneity and plasticity. PLoS ONE 2011, 6, e16186. [Google Scholar] [CrossRef]
  53. Xu, S.; Yang, Y.; Dong, L.; Qiu, W.; Yang, L.; Wang, X.; Liu, L. Construction and characteristics of an E-cadherin-related three-dimensional suspension growth model of ovarian cancer. Sci. Rep. 2014, 4, 5646. [Google Scholar] [CrossRef] [PubMed]
  54. Reddy, P.; Liu, L.; Ren, C.; Lindgren, P.; Boman, K.; Shen, Y.; Lundin, E.; Ottander, U.; Rytinki, M.; Liu, K. Formation of E-cadherin-mediated cell-cell adhesion activates AKT and mitogen activated protein kinase via phosphatidylinositol 3 kinase and ligand-independent activation of epidermal growth factor receptor in ovarian cancer cells. Mol. Endocrinol. 2005, 19, 2564–2578. [Google Scholar] [CrossRef] [PubMed]
  55. Huber, M.A.; Kraut, N.; Beug, H. Molecular requirements for epithelial-mesenchymal transition during tumor progression. Curr. Opin. Cell Biol. 2005, 17, 548–558. [Google Scholar] [CrossRef] [PubMed]
  56. Dong, L.L.; Liu, L.; Ma, C.H.; Li, J.S.; Du, C.; Xu, S.; Han, L.H.; Li, L.; Wang, X.W. E-cadherin promotes proliferation of human ovarian cancer cells in vitro via activating MEK/ERK pathway. Acta Pharmacol. Sin. 2012, 33, 817–822. [Google Scholar] [CrossRef] [PubMed]
  57. Davis, M.A.; Ireton, R.C.; Reynolds, A.B. A core function for p120-catenin in cadherin turnover. J. Cell Biol. 2003, 163, 525–534. [Google Scholar] [CrossRef] [PubMed]
  58. Maretzky, T.; Reiss, K.; Ludwig, A.; Buchholz, J.; Scholz, F.; Proksch, E.; de Strooper, B.; Hartmann, D.; Saftig, P. ADAM10 mediates E-cadherin shedding and regulates epithelial cell-cell adhesion, migration, and β-catenin translocation. Proc. Natl. Acad. Sci. USA 2005, 102, 9182–9187. [Google Scholar] [CrossRef] [PubMed]
  59. Noe, V.; Fingleton, B.; Jacobs, K.; Crawford, H.C.; Vermeulen, S.; Steelant, W.; Bruyneel, E.; Matrisian, L.M.; Mareel, M. Release of an invasion promoter E-cadherin fragment by matrilysin and stromelysin-1. J. Cell Sci. 2001, 114, 111–118. [Google Scholar] [PubMed]
  60. Marambaud, P.; Shioi, J.; Serban, G.; Georgakopoulos, A.; Sarner, S.; Nagy, V.; Baki, L.; Wen, P.; Efthimiopoulos, S.; Shao, Z.; et al. A presenilin-1/γ-secretase cleavage releases the E-cadherin intracellular domain and regulates disassembly of adherens junctions. EMBO J. 2002, 21, 1948–1956. [Google Scholar] [CrossRef] [PubMed]
  61. Gogali, A.; Charalabopoulos, K.; Zampira, I.; Konstantinidis, A.K.; Tachmazoglou, F.; Daskalopoulos, G.; Constantopoulos, S.H.; Dalavanga, Y. Soluble adhesion molecules E-cadherin, intercellular adhesion molecule-1, and E-selectin as lung cancer biomarkers. Chest 2010, 138, 1173–1179. [Google Scholar] [CrossRef] [PubMed]
  62. Cowden Dahl, K.D.; Symowicz, J.; Ning, Y.; Gutierrez, E.; Fishman, D.A.; Adley, B.P.; Stack, M.S.; Hudson, L.G. Matrix metalloproteinase 9 is a mediator of epidermal growth factor-dependent E-cadherin loss in ovarian carcinoma cells. Cancer Res. 2008, 68, 4606–4613. [Google Scholar] [CrossRef] [PubMed]
  63. Symowicz, J.; Adley, B.P.; Gleason, K.J.; Johnson, J.J.; Ghosh, S.; Fishman, D.A.; Hudson, L.G.; Stack, M.S. Engagement of collagen-binding integrins promotes matrix metalloproteinase-9-dependent E-cadherin ectodomain shedding in ovarian carcinoma cells. Cancer Res. 2007, 67, 2030–2039. [Google Scholar] [CrossRef] [PubMed]
  64. Trillsch, F.; Kuerti, S.; Eulenburg, C.; Burandt, E.; Woelber, L.; Prieske, K.; Eylmann, K.; Oliveira-Ferrer, L.; Milde-Langosch, K.; Mahner, S. E-Cadherin fragments as potential mediators for peritoneal metastasis in advanced epithelial ovarian cancer. Br. J. Cancer 2016, 114, 213–220. [Google Scholar] [CrossRef] [PubMed]
  65. De Boer, C.J.; van, D.E.; van, K.H.; van Jansen Rhijn, C.M.; Warnaar, S.O.; Fleuren, G.J.; Litvinov, S.V. Changing roles of cadherins and catenins during progression of squamous intraepithelial lesions in the uterine cervix. Am. J. Pathol. 1999, 155, 505–515. [Google Scholar] [CrossRef]
  66. Bailey, T.; Biddlestone, L.; Shepherd, N.; Barr, H.; Warner, P.; Jankowski, J. Altered cadherin and catenin complexes in the Barrett’s esophagus-dysplasia-adenocarcinoma sequence: Correlation with disease progression and dedifferentiation. Am. J. Pathol. 1998, 152, 135–144. [Google Scholar] [PubMed]
  67. Peralta, S.A.; Knudsen, K.A.; Salazar, H.; Han, A.C.; Keshgegian, A.A. P-cadherin expression in breast carcinoma indicates poor survival. Cancer 1999, 86, 1263–1272. [Google Scholar]
  68. Hardy, R.G.; Tselepis, C.; Hoyland, J.; Wallis, Y.; Pretlow, T.P.; Talbot, I.; Sanders, D.S.; Matthews, G.; Morton, D.; Jankowski, J.A. Aberrant P-cadherin expression is an early event in hyperplastic and dysplastic transformation in the colon. Gut 2002, 50, 513–519. [Google Scholar] [CrossRef] [PubMed]
  69. Patel, I.S.; Madan, P.; Getsios, S.; Bertrand, M.A.; MacCalman, C.D. Cadherin switching in ovarian cancer progression. Int. J. Cancer 2003, 106, 172–177. [Google Scholar] [CrossRef] [PubMed]
  70. Cheung, L.W.; Leung, P.C.; Wong, A.S. Cadherin switching and activation of p120 catenin signaling are mediators of gonadotropin-releasing hormone to promote tumor cell migration and invasion in ovarian cancer. Oncogene 2010, 29, 2427–2440. [Google Scholar] [CrossRef] [PubMed]
  71. Cheung, L.W.; Mak, A.S.; Cheung, A.N.; Ngan, H.Y.; Leung, P.C.; Wong, A.S. P-cadherin cooperates with insulin-like growth factor-1 receptor to promote metastatic signaling of gonadotropin-releasing hormone in ovarian cancer via p120 catenin. Oncogene 2011, 30, 2964–2974. [Google Scholar] [CrossRef] [PubMed]
  72. Cheung, L.W.; Yung, S.; Chan, T.M.; Leung, P.C.; Wong, A.S. Targeting gonadotropin-releasing hormone receptor inhibits the early step of ovarian cancer metastasis by modulating tumor-mesothelial adhesion. Mol. Ther. 2013, 21, 78–90. [Google Scholar] [CrossRef] [PubMed]
  73. Usui, A.; Ko, S.Y.; Barengo, N.; Naora, H. P-cadherin promotes ovarian cancer dissemination through tumor cell aggregation and tumor-peritoneum interactions. Mol. Cancer Res. 2014, 12, 504–513. [Google Scholar] [CrossRef] [PubMed]
  74. Ip, C.K.; Yung, S.; Chan, T.M.; Tsao, S.W.; Wong, A.S. p70 S6 kinase drives ovarian cancer metastasis through multicellular spheroid-peritoneum interaction and P-cadherin/β1 integrin signaling activation. Oncotarget 2014, 5, 9133–9149. [Google Scholar] [PubMed]
  75. Barczyk, M.; Carracedo, S.; Gullberg, D. Integrins. Cell Tissue Res. 2010, 339, 269–280. [Google Scholar] [CrossRef]
  76. Ruoslahti, E. RGD and other recognition sequences for integrins. Annu. Rev. Cell Biol. 1996, 12, 697–715. [Google Scholar] [CrossRef] [PubMed]
  77. Zhao, X.; Guan, J.L. Focal adhesion kinase and its signaling pathways in cell migration and angiogenesis. Adv. Drug Deliv. Rev. 2011, 63, 610–615. [Google Scholar] [CrossRef] [PubMed]
  78. Schlaepfer, D.D.; Hanks, S.K.; Hunter, T.; van der, G.P. Integrin-mediated signal transduction linked to Ras pathway by GRB2 binding to focal adhesion kinase. Nature 1994, 372, 786–791. [Google Scholar] [CrossRef] [PubMed]
  79. Mitra, S.K.; Schlaepfer, D.D. Integrin-regulated FAK-Src signaling in normal and cancer cells. Curr. Opin. Cell Biol. 2006, 18, 516–523. [Google Scholar] [CrossRef] [PubMed]
  80. Heyman, L.; Leroy-Dudal, J.; Fernandes, J.; Seyer, D.; Dutoit, S.; Carreiras, F. Mesothelial vitronectin stimulates migration of ovarian cancer cells. Cell Biol. Int. 2010, 34, 493–502. [Google Scholar] [CrossRef] [PubMed]
  81. Sawada, K.; Mitra, A.K.; Radjabi, A.R.; Bhaskar, V.; Kistner, E.O.; Tretiakova, M.; Jagadeeswaran, S.; Montag, A.; Becker, A.; Kenny, H.A.; et al. Loss of E-cadherin promotes ovarian cancer metastasis via α 5-integrin, which is a therapeutic target. Cancer Res. 2008, 68, 2329–2339. [Google Scholar] [CrossRef] [PubMed]
  82. Ahmed, N.; Pansino, F.; Clyde, R.; Murthi, P.; Quinn, M.A.; Rice, G.E.; Agrez, M.V.; Mok, S.; Baker, M.S. Overexpression of αvβ6 integrin in serous epithelial ovarian cancer regulates extracellular matrix degradation via the plasminogen activation cascade. Carcinogenesis 2002, 23, 237–244. [Google Scholar] [CrossRef] [PubMed]
  83. Carduner, L.; Picot, C.R.; Leroy-Dudal, J.; Blay, L.; Kellouche, S.; Carreiras, F. Cell cycle arrest or survival signaling through αv integrins, activation of PKC and ERK1/2 lead to anoikis resistance of ovarian cancer spheroids. Exp. Cell Res. 2014, 320, 329–342. [Google Scholar] [CrossRef] [PubMed]
  84. Carduner, L.; Leroy-Dudal, J.; Picot, C.R.; Gallet, O.; Carreiras, F.; Kellouche, S. Ascites-induced shift along epithelial-mesenchymal spectrum in ovarian cancer cells: Enhancement of their invasive behavior partly dependant on αv integrins. Clin. Exp. Metastasis 2014, 31, 675–688. [Google Scholar] [CrossRef] [PubMed]
  85. Cho, A.; Howell, V.M.; Colvin, E.K. The Extracellular matrix in epithelial ovarian cancer—A piece of a Puzzle. Front. Oncol. 2015, 5, 245. [Google Scholar] [PubMed]
  86. Kenny, H.A.; Chiang, C.Y.; White, E.A.; Schryver, E.M.; Habis, M.; Romero, I.L.; Ladanyi, A.; Penicka, C.V.; George, J.; Matlin, K.; et al. Mesothelial cells promote early ovarian cancer metastasis through fibronectin secretion. J. Clin. Investig. 2014, 124, 4614–4628. [Google Scholar] [CrossRef] [PubMed]
  87. Yousif, N.G. Fibronectin promotes migration and invasion of ovarian cancer cells through up-regulation of FAK-PI3K/Akt pathway. Cell Biol. Int. 2014, 38, 85–91. [Google Scholar] [CrossRef] [PubMed]
  88. Franke, F.E.; Von, G.R.; Zygmunt, M.; Munstedt, K. Association between fibronectin expression and prognosis in ovarian carcinoma. Anticancer Res. 2003, 23, 4261–4267. [Google Scholar] [PubMed]
  89. Desgrosellier, J.S.; Cheresh, D.A. Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9–22. [Google Scholar] [CrossRef] [PubMed]
  90. Ivaska, J.; Heino, J. Cooperation between integrins and growth factor receptors in signaling and endocytosis. Annu. Rev. Cell Dev. Biol. 2011, 27, 291–320. [Google Scholar] [CrossRef] [PubMed]
  91. Regad, T. Targeting RTK signaling pathways in cancer. Cancers 2015, 7, 1758–1784. [Google Scholar] [CrossRef] [PubMed]
  92. Jiao, Y.; Ou, W.; Meng, F.; Zhou, H.; Wang, A. Targeting HSP90 in ovarian cancers with multiple receptor tyrosine kinase coactivation. Mol. Cancer 2011, 10, 125. [Google Scholar] [CrossRef] [PubMed]
  93. Citri, A.; Yarden, Y. EGF-ERBB signalling: Towards the systems level. Nat. Rev. Mol. Cell Biol. 2006, 7, 505–516. [Google Scholar] [CrossRef] [PubMed]
  94. Moro, L.; Venturino, M.; Bozzo, C.; Silengo, L.; Altruda, F.; Beguinot, L.; Tarone, G.; Defilippi, P. Integrins induce activation of EGF receptor: Role in MAP kinase induction and adhesion-dependent cell survival. EMBO J. 1998, 17, 6622–6632. [Google Scholar] [CrossRef] [PubMed]
  95. Mariotti, A.; Kedeshian, P.A.; Dans, M.; Curatola, A.M.; Gagnoux-Palacios, L.; Giancotti, F.G. EGF-R signaling through Fyn kinase disrupts the function of integrin α6β4 at hemidesmosomes: Role in epithelial cell migration and carcinoma invasion. J. Cell Biol. 2001, 155, 447–458. [Google Scholar] [CrossRef] [PubMed]
  96. Moro, L.; Dolce, L.; Cabodi, S.; Bergatto, E.; Boeri, E.E.; Smeriglio, M.; Turco, E.; Retta, S.F.; Giuffrida, M.G.; Venturino, M.; et al. Integrin-induced epidermal growth factor (EGF) receptor activation requires c-Src and p130Cas and leads to phosphorylation of specific EGF receptor tyrosines. J. Biol. Chem. 2002, 277, 9405–9414. [Google Scholar] [CrossRef] [PubMed]
  97. Cabodi, S.; Moro, L.; Bergatto, E.; Boeri, E.E.; Di, S.P.; Turco, E.; Tarone, G.; Defilippi, P. Integrin regulation of epidermal growth factor (EGF) receptor and of EGF-dependent responses. Biochem. Soc. Trans. 2004, 32, 438–442. [Google Scholar] [CrossRef] [PubMed]
  98. Gui, T.; Shen, K. The epidermal growth factor receptor as a therapeutic target in epithelial ovarian cancer. Cancer Epidemiol. 2012, 36, 490–496. [Google Scholar] [CrossRef] [PubMed]
  99. Lafky, J.M.; Wilken, J.A.; Baron, A.T.; Maihle, N.J. Clinical implications of the ErbB/epidermal growth factor (EGF) receptor family and its ligands in ovarian cancer. Biochim. Biophys. Acta 2008, 1785, 232–265. [Google Scholar] [CrossRef] [PubMed]
  100. Alberti, C.; Pinciroli, P.; Valeri, B.; Ferri, R.; Ditto, A.; Umezawa, K.; Sensi, M.; Canevari, S.; Tomassetti, A. Ligand-dependent EGFR activation induces the co-expression of IL-6 and PAI-1 via the NFκB pathway in advanced-stage epithelial ovarian cancer. Oncogene 2012, 31, 4139–4149. [Google Scholar] [CrossRef] [PubMed]
  101. Alper, O.; Bergmann-Leitner, E.S.; Bennett, T.A.; Hacker, N.F.; Stromberg, K.; Stetler-Stevenson, W.G. Epidermal growth factor receptor signaling and the invasive phenotype of ovarian carcinoma cells. J. Natl. Cancer Inst. 2001, 93, 1375–1384. [Google Scholar] [CrossRef] [PubMed]
  102. Ning, Y.; Zeineldin, R.; Liu, Y.; Rosenberg, M.; Stack, M.S.; Hudson, L.G. Down-regulation of integrin α2 surface expression by mutant epidermal growth factor receptor (EGFRvIII) induces aberrant cell spreading and focal adhesion formation. Cancer Res. 2005, 65, 9280–9286. [Google Scholar] [CrossRef] [PubMed]
  103. Comoglio, P.M. Pathway specificity for Met signalling. Nat. Cell Biol. 2001, 3, E161–E162. [Google Scholar] [CrossRef] [PubMed]
  104. Sawada, K.; Radjabi, A.R.; Shinomiya, N.; Kistner, E.; Kenny, H.; Becker, A.R.; Turkyilmaz, M.A.; Salgia, R.; Yamada, S.D.; Vande Woude, G.F.; et al. c-Met overexpression is a prognostic factor in ovarian cancer and an effective target for inhibition of peritoneal dissemination and invasion. Cancer Res. 2007, 67, 1670–1679. [Google Scholar] [CrossRef] [PubMed]
  105. Mitra, A.K.; Sawada, K.; Tiwari, P.; Mui, K.; Gwin, K.; Lengyel, E. Ligand-independent activation of c-Met by fibronectin and α5β1-integrin regulates ovarian cancer invasion and metastasis. Oncogene 2011, 30, 1566–1576. [Google Scholar] [CrossRef] [PubMed]
  106. Kwon, Y.; Smith, B.D.; Zhou, Y.; Kaufman, M.D.; Godwin, A.K. Effective inhibition of c-MET-mediated signaling, growth and migration of ovarian cancer cells is influenced by the ovarian tissue microenvironment. Oncogene 2015, 34, 144–153. [Google Scholar] [CrossRef] [PubMed]
  107. Moran-Jones, K.; Brown, L.M.; Samimi, G. INC280, an orally available small molecule inhibitor of c-MET, reduces migration and adhesion in ovarian cancer cell models. Sci. Rep. 2015, 5, 11749. [Google Scholar] [CrossRef] [PubMed]
  108. Ferrara, N.; Hillan, K.J.; Gerber, H.P.; Novotny, W. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer. Nat. Rev. Drug Discov. 2004, 3, 391–400. [Google Scholar] [CrossRef] [PubMed]
  109. Mu, J.; Abe, Y.; Tsutsui, T.; Yamamoto, N.; Tai, X.G.; Niwa, O.; Tsujimura, T.; Sato, B.; Terano, H.; Fujiwara, H.; et al. Inhibition of growth and metastasis of ovarian carcinoma by administering a drug capable of interfering with vascular endothelial growth factor activity. Jpn. J. Cancer Res. 1996, 87, 963–971. [Google Scholar] [CrossRef] [PubMed]
  110. Yamamoto, S.; Konishi, I.; Mandai, M.; Kuroda, H.; Komatsu, T.; Nanbu, K.; Sakahara, H.; Mori, T. Expression of vascular endothelial growth factor (VEGF) in epithelial ovarian neoplasms: Correlation with clinicopathology and patient survival, and analysis of serum VEGF levels. Br. J. Cancer 1997, 76, 1221–1227. [Google Scholar] [CrossRef] [PubMed]
  111. Serini, G.; Napione, L.; Arese, M.; Bussolino, F. Besides adhesion: New perspectives of integrin functions in angiogenesis. Cardiovasc. Res. 2008, 78, 213–222. [Google Scholar] [CrossRef] [PubMed]
  112. Kim, T.J.; Landen, C.N.; Lin, Y.G.; Mangala, L.S.; Lu, C.; Nick, A.M.; Stone, R.L.; Merritt, W.M.; rmaiz-Pena, G.; Jennings, N.B.; et al. Combined anti-angiogenic therapy against VEGF and integrin αvβ3 in an orthotopic model of ovarian cancer. Cancer Biol. Ther. 2009, 8, 2263–2272. [Google Scholar] [CrossRef] [PubMed]
  113. Landen, C.N.; Kim, T.J.; Lin, Y.G.; Merritt, W.M.; Kamat, A.A.; Han, L.Y.; Spannuth, W.A.; Nick, A.M.; Jennnings, N.B.; Kinch, M.S.; et al. Tumor-selective response to antibody-mediated targeting of αvβ3 integrin in ovarian cancer. Neoplasia 2008, 10, 1259–1267. [Google Scholar] [CrossRef] [PubMed]
  114. Brown, M.; Black, J.R.; Sharma, R.; Stebbing, J.; Pinato, D.J. Gene of the month: Axl. J. Clin. Pathol. 2016, 69, 391–397. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, Z.; Lee, J.C.; Lin, L.; Olivas, V.; Au, V.; LaFramboise, T.; bdel-Rahman, M.; Wang, X.; Levine, A.D.; Rho, J.K.; et al. Activation of the AXL kinase causes resistance to EGFR-targeted therapy in lung cancer. Nat. Genet. 2012, 44, 852–860. [Google Scholar] [CrossRef] [PubMed]
  116. Byers, L.A.; Diao, L.; Wang, J.; Saintigny, P.; Girard, L.; Peyton, M.; Shen, L.; Fan, Y.; Giri, U.; Tumula, P.K.; et al. An epithelial-mesenchymal transition gene signature predicts resistance to EGFR and PI3K inhibitors and identifies Axl as a therapeutic target for overcoming EGFR inhibitor resistance. Clin. Cancer Res. 2013, 19, 279–290. [Google Scholar] [CrossRef] [PubMed]
  117. Linger, R.M.; Keating, A.K.; Earp, H.S.; Graham, D.K. TAM receptor tyrosine kinases: Biologic functions, signaling, and potential therapeutic targeting in human cancer. Adv. Cancer Res. 2008, 100, 35–83. [Google Scholar] [PubMed]
  118. Korshunov, V.A. Axl-dependent signalling: A clinical update. Clin. Sci. 2012, 122, 361–368. [Google Scholar] [CrossRef] [PubMed]
  119. Sun, W.; Fujimoto, J.; Tamaya, T. Coexpression of Gas6/Axl in human ovarian cancers. Oncology 2004, 66, 450–457. [Google Scholar] [CrossRef] [PubMed]
  120. Buehler, M.; Tse, B.; Leboucq, A.; Jacob, F.; Caduff, R.; Fink, D.; Goldstein, D.R.; Heinzelmann-Schwarz, V. Meta-analysis of microarray data identifies GAS6 expression as an independent predictor of poor survival in ovarian cancer. BioMed Res. Int. 2013, 2013, 238284. [Google Scholar] [CrossRef] [PubMed]
  121. tRankin, E.B.; Fuh, K.C.; Taylor, T.E.; Krieg, A.J.; Musser, M.; Yuan, J.; Wei, K.; Kuo, C.J.; Longacre, T.A.; Giaccia, A.J. Axl is an essential factor and therapeutic target for metastatic ovarian cancer. Cancer Res. 2010, 70, 7570–7579. [Google Scholar] [CrossRef] [PubMed]
  122. Rea, K.; Pinciroli, P.; Sensi, M.; Alciato, F.; Bisaro, B.; Lozneanu, L.; Raspagliesi, F.; Centritto, F.; Cabodi, S.; Defilippi, P.; et al. Novel Axl-driven signaling pathway and molecular signature characterize high-grade ovarian cancer patients with poor clinical outcome. Oncotarget 2015, 6, 30859–30875. [Google Scholar] [PubMed]
  123. Acquati, F.; Morelli, C.; Cinquetti, R.; Bianchi, M.G.; Porrini, D.; Varesco, L.; Gismondi, V.; Rocchetti, R.; Talevi, S.; Possati, L.; et al. Cloning and characterization of a senescence inducing and class II tumor suppressor gene in ovarian carcinoma at chromosome region 6q27. Oncogene 2001, 20, 980–988. [Google Scholar] [CrossRef] [PubMed]
  124. Acquati, F.; Possati, L.; Ferrante, L.; Campomenosi, P.; Talevi, S.; Bardelli, S.; Margiotta, C.; Russo, A.; Bortoletto, E.; Rocchetti, R.; et al. Tumor and metastasis suppression by the human RNASET2 gene. Int. J. Oncol. 2005, 26, 1159–1168. [Google Scholar] [CrossRef] [PubMed]
  125. Acquati, F.; Lualdi, M.; Bertilaccio, S.; Monti, L.; Turconi, G.; Fabbri, M.; Grimaldi, A.; Anselmo, A.; Inforzato, A.; Collotta, A.; et al. Loss of function of Ribonuclease T2, an ancient and phylogenetically conserved RNase, plays a crucial role in ovarian tumorigenesis. Proc. Natl. Acad. Sci. USA 2013, 110, 8140–8145. [Google Scholar] [CrossRef] [PubMed]
  126. Lualdi, M.; Pedrini, E.; Rea, K.; Monti, L.; Scaldaferri, D.; Gariboldi, M.; Camporeale, A.; Ghia, P.; Monti, E.; Tomassetti, A.; et al. Pleiotropic modes of action in tumor cells of RNASET2, an evolutionary highly conserved extracellular RNase. Oncotarget 2015, 6, 7851–7865. [Google Scholar] [CrossRef] [PubMed]
  127. Smirnoff, P.; Roiz, L.; Angelkovitch, B.; Schwartz, B.; Shoseyov, O. A recombinant human RNASET2 glycoprotein with antitumorigenic and antiangiogenic characteristics: Expression, purification, and characterization. Cancer 2006, 107, 2760–2769. [Google Scholar] [CrossRef] [PubMed]
  128. Nesiel-Nuttman, L.; Schwartz, B.; Shoseyov, O. Human recombinant truncated RNASET2, devoid of RNase activity; A potential cancer therapeutic agent. Oncotarget 2014, 5, 11464–11478. [Google Scholar] [CrossRef] [PubMed]
  129. Yin, F.; Liu, L.; Liu, X.; Li, G.; Zheng, L.; Li, D.; Wang, Q.; Zhang, W.; Li, L. Downregulation of tumor suppressor gene ribonuclease T2 and gametogenetin binding protein 2 is associated with drug resistance in ovarian cancer. Oncol. Rep. 2014, 32, 362–372. [Google Scholar] [CrossRef] [PubMed]
  130. Eke, I.; Cordes, N. Focal adhesion signaling and therapy resistance in cancer. Semin. Cancer Biol. 2015, 31, 65–75. [Google Scholar] [CrossRef] [PubMed]
  131. Seguin, L.; Desgrosellier, J.S.; Weis, S.M.; Cheresh, D.A. Integrins and cancer: Regulators of cancer stemness, metastasis, and drug resistance. Trends Cell Biol. 2015, 25, 234–240. [Google Scholar] [CrossRef] [PubMed]
  132. Lanzardo, S.; Conti, L.; Brioschi, C.; Bartolomeo, M.P.; Arosio, D.; Belvisi, L.; Manzoni, L.; Maiocchi, A.; Maisano, F.; Forni, G. A new optical imaging probe targeting αvβ3 integrin in glioblastoma xenografts. Contrast Media Mol. Imaging 2011, 6, 449–458. [Google Scholar] [CrossRef] [PubMed]
  133. Manzoni, L.; Belvisi, L.; Arosio, D.; Bartolomeo, M.P.; Bianchi, A.; Brioschi, C.; Buonsanti, F.; Cabella, C.; Casagrande, C.; Civera, M.; et al. Synthesis of Gd and 68Ga complexes in conjugation with a conformationally optimized RGD sequence as potential MRI and PET tumor-imaging probes. ChemMedChem 2012, 7, 1084–1093. [Google Scholar] [CrossRef] [PubMed]
  134. Pilkington-Miksa, M.; Arosio, D.; Battistini, L.; Belvisi, L.; De, M.M.; Vasile, F.; Burreddu, P.; Carta, P.; Rassu, G.; Perego, P.; et al. Design, synthesis, and biological evaluation of novel cRGD-paclitaxel conjugates for integrin-assisted drug delivery. Bioconjug. Chem. 2012, 23, 1610–1622. [Google Scholar] [CrossRef] [PubMed]
  135. Colombo, R.; Mingozzi, M.; Belvisi, L.; Arosio, D.; Piarulli, U.; Carenini, N.; Perego, P.; Zaffaroni, N.; de, C.M.; Castiglioni, V.; et al. Synthesis and biological evaluation (in vitro and in vivo) of cyclic arginine-glycine-aspartate (RGD) peptidomimetic-paclitaxel conjugates targeting integrin αvβ3. J. Med. Chem. 2012, 55, 10460–10474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Sawada, K.; Ohyagi-Hara, C.; Kimura, T.; Morishige, K. Integrin inhibitors as a therapeutic agent for ovarian cancer. J. Oncol. 2012, 2012, 915140. [Google Scholar] [CrossRef] [PubMed]
  137. Stupp, R.; Hegi, M.E.; Gorlia, T.; Erridge, S.C.; Perry, J.; Hong, Y.K.; Aldape, K.D.; Lhermitte, B.; Pietsch, T.; Grujicic, D.; et al. Cilengitide combined with standard treatment for patients with newly diagnosed glioblastoma with methylated MGMT promoter (CENTRIC EORTC 26071–22072 study): A multicentre, randomised, open-label, phase 3 trial. Lancet Oncol. 2014, 15, 1100–1108. [Google Scholar] [CrossRef]
  138. Merck Press Release on Cilengitide Studies. Available online: http://www.merck.de/de/press/extNewsDetail.html?newsId=C47977D13865FCB9C1257B1D001EF9CA&newsType=1 (accessed on 30 June 2016).
  139. Arosio, D.; Casagrande, C. Advancement in integrin facilitated drug delivery. Adv. Drug Deliv. Rev. 2016, 97, 111–143. [Google Scholar] [CrossRef] [PubMed]
  140. Blaschuk, O.W. Discovery and development of N-cadherin antagonists. Cell Tissue Res. 2012, 348, 309–313. [Google Scholar] [CrossRef] [PubMed]
  141. Augustine, C.K.; Yoshimoto, Y.; Gupta, M.; Zipfel, P.A.; Selim, M.A.; Febbo, P.; Pendergast, A.M.; Peters, W.P.; Tyler, D.S. Targeting N-cadherin enhances antitumor activity of cytotoxic therapies in melanoma treatment. Cancer Res. 2008, 68, 3777–3784. [Google Scholar] [CrossRef] [PubMed]
  142. Beasley, G.M.; Riboh, J.C.; Augustine, C.K.; Zager, J.S.; Hochwald, S.N.; Grobmyer, S.R.; Peterson, B.; Royal, R.; Ross, M.I.; Tyler, D.S. Prospective Multicenter Phase II trial of systemic ADH-1 in combination with melphalan via isolated limb infusion in patients with advanced extremity melanoma. J. Clin. Oncol. 2011, 29, 1210–1215. [Google Scholar] [CrossRef] [PubMed]
  143. Perotti, A.; Sessa, C.; Mancuso, A.; Noberasco, C.; Cresta, S.; Locatelli, A.; Carcangiu, M.L.; Passera, K.; Braghetti, A.; Scaramuzza, D.; et al. Clinical and pharmacological phase I evaluation of Exherin (ADH-1), a selective anti-N-cadherin peptide in patients with N-cadherin-expressing solid tumours. Ann. Oncol. 2009, 20, 741–745. [Google Scholar] [CrossRef] [PubMed]
  144. Turley, R.S.; Tokuhisa, Y.; Toshimitsu, H.; Lidsky, M.E.; Padussis, J.C.; Fontanella, A.; Deng, W.; Augustine, C.K.; Beasley, G.M.; Davies, M.A.; et al. Targeting N-cadherin increases vascular permeability and differentially activates AKT in melanoma. Ann. Surg. 2015, 261, 368–377. [Google Scholar] [CrossRef] [PubMed]
  145. Doro, F.; Colombo, C.; Alberti, C.; Arosio, D.; Belvisi, L.; Casagrande, C.; Fanelli, R.; Manzoni, L.; Parisini, E.; Piarulli, U.; et al. Computational design of novel peptidomimetic inhibitors of cadherin homophilic interactions. Org. Biomol. Chem. 2015, 13, 2570–2573. [Google Scholar] [CrossRef] [PubMed]
  146. Nardone, V.; Lucarelli, A.P.; Dalle, V.A.; Fanelli, R.; Tomassetti, A.; Belvisi, L.; Civera, M.; Parisini, E. Crystal structure of human E-Cadherin-EC1EC2 in complex with a peptidomimetic competitive inhibitor of cadherin homophilic interaction. J. Med. Chem. 2016, 59, 5089–5094. [Google Scholar] [CrossRef] [PubMed]
  147. Dechantsreiter, M.A.; Planker, E.; Matha, B.; Lohof, E.; Holzemann, G.; Jonczyk, A.; Goodman, S.L.; Kessler, H. N-Methylated cyclic RGD peptides as highly active and selective αVβ3 integrin antagonists. J. Med. Chem. 1999, 42, 3033–3040. [Google Scholar] [CrossRef] [PubMed]
  148. Arap, W.; Pasqualini, R.; Ruoslahti, E. Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model. Science 1998, 279, 377–380. [Google Scholar] [CrossRef] [PubMed]
  149. Manzoni, L.; Belvisi, L.; Arosio, D.; Civera, M.; Pilkington-Miksa, M.; Potenza, D.; Caprini, A.; Araldi, E.M.; Monferini, E.; Mancino, M.; et al. Cyclic RGD-containing functionalized azabicycloalkane peptides as potent integrin antagonists for tumor targeting. ChemMedChem 2009, 4, 615–632. [Google Scholar] [CrossRef] [PubMed]
  150. Hou, L.; Zhao, X.; Wang, P.; Ning, Q.; Meng, M.; Liu, C. Antitumor activity of antimicrobial peptides containing CisoDGRC in CD13 negative breast cancer cells. PLoS ONE 2013, 8, e53491. [Google Scholar] [CrossRef] [PubMed]
  151. Panzeri, S.; Zanella, S.; Arosio, D.; Vahdati, L.; Dal, C.A.; Pignataro, L.; Paolillo, M.; Schinelli, S.; Belvisi, L.; Gennari, C.; et al. Cyclic isoDGR and RGD peptidomimetics containing bifunctional diketopiperazine scaffolds are integrin antagonists. Chemistry 2015, 21, 6265–6271. [Google Scholar] [CrossRef] [PubMed]
  152. Li, S.; Gray, B.P.; McGuire, M.J.; Brown, K.C. Synthesis and biological evaluation of a peptide-paclitaxel conjugate which targets the integrin αvβ6. Bioorg. Med. Chem. 2011, 19, 5480–5489. [Google Scholar] [CrossRef] [PubMed]
  153. Shintani, Y.; Fukumoto, Y.; Chaika, N.; Grandgenett, P.M.; Hollingsworth, M.A.; Wheelock, M.J.; Johnson, K.R. ADH-1 suppresses N-cadherin-dependent pancreatic cancer progression. Int. J. Cancer 2008, 122, 71–77. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of (a) E- and (b) P-cadh-associated signaling cascades activated in EOCs (Epithelial Ovarian Cancers). The arrows indicate: green, protein-protein interaction, black, signaling cascades; discontinuous, possible signaling cascade; red, cellular effects. The abbreviation used are: cadh, cadherin; IGF-1R, Insulin Growth Factor Receptor 1; ERK, Extracellular signal-Regulated Kinase; MEK, MAP or ERK kinase; PI3K, phosphoinositide 3-kinase; p120, p120 catenin. Rho, Rac and CDC42 are Rho-GTPases.
Figure 1. Schematic representation of (a) E- and (b) P-cadh-associated signaling cascades activated in EOCs (Epithelial Ovarian Cancers). The arrows indicate: green, protein-protein interaction, black, signaling cascades; discontinuous, possible signaling cascade; red, cellular effects. The abbreviation used are: cadh, cadherin; IGF-1R, Insulin Growth Factor Receptor 1; ERK, Extracellular signal-Regulated Kinase; MEK, MAP or ERK kinase; PI3K, phosphoinositide 3-kinase; p120, p120 catenin. Rho, Rac and CDC42 are Rho-GTPases.
Ijms 17 01387 g001
Figure 2. Schematic representation of the the signaling cascades activated in cancer cells by the cross-talk between integrins and RTKs. The arrays indicate: black, signaling cascades; yellow, phosphorylation; discontinuous, possible signaling cascades; blue, trafficking; red, effect. The abbreviations are: RTK, Receptor Tyrosine Kinase; ECM, Extra Cellular Matrix, MAPK, Mitogen Activated Protein Kinase, FAK, Focal Adhesion Kinase; src, Sarcoma viral oncogene; ERK, Extracellular signal-Regulated Kinase.
Figure 2. Schematic representation of the the signaling cascades activated in cancer cells by the cross-talk between integrins and RTKs. The arrays indicate: black, signaling cascades; yellow, phosphorylation; discontinuous, possible signaling cascades; blue, trafficking; red, effect. The abbreviations are: RTK, Receptor Tyrosine Kinase; ECM, Extra Cellular Matrix, MAPK, Mitogen Activated Protein Kinase, FAK, Focal Adhesion Kinase; src, Sarcoma viral oncogene; ERK, Extracellular signal-Regulated Kinase.
Ijms 17 01387 g002
Table 1. Integrins or cadherins expressed on EOC cells and their suitable ligands.
Table 1. Integrins or cadherins expressed on EOC cells and their suitable ligands.
IntegrinsLigandsChemical ScaffoldTumor Cell Model 1Reference
αvβ3cRGDfV 2CyclopentapeptideGBM[147]
RGD4CCyclopentapeptideBC[148]
cAbaRGDAzabicycloalkaneEOC[149]
(DKP)-RGDDichetopiperazineEOC[135]
CisoDGRCDAK 22-mer peptideBC[150]
Cyclo[DKP-isoDGR]Dichetopiperazine/CDAKGBM[151]
α5β3H2009.120-mer peptideNSCL-C[152]
Cadherins
N-cadhN-Ac-CHAVC-NH2 3Disulphade-linked cyclic peptidePC[153]
N- and E-cadhCompound 3Benzyl ringEOC[145]
1 Only ligands tested on cancer cells are reported. The abbreviations are: GBM, Glioblastoma; BC, Breast Cancer; NSCL-C Non-Small Cell Lung cancer; PC, Pancreatic cancer; 2 Cilengitide (Merck, Darmstadt, Germany). It also binds to α5β3 integrin; 3 ADH-1 or Exherin.

Share and Cite

MDPI and ACS Style

Roggiani, F.; Mezzanzanica, D.; Rea, K.; Tomassetti, A. Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells. Int. J. Mol. Sci. 2016, 17, 1387. https://doi.org/10.3390/ijms17091387

AMA Style

Roggiani F, Mezzanzanica D, Rea K, Tomassetti A. Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells. International Journal of Molecular Sciences. 2016; 17(9):1387. https://doi.org/10.3390/ijms17091387

Chicago/Turabian Style

Roggiani, Francesca, Delia Mezzanzanica, Katia Rea, and Antonella Tomassetti. 2016. "Guidance of Signaling Activations by Cadherins and Integrins in Epithelial Ovarian Cancer Cells" International Journal of Molecular Sciences 17, no. 9: 1387. https://doi.org/10.3390/ijms17091387

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop