Open access

Molecular Mechanisms of Metastasis: Epithelial-Mesenchymal Transition, Anoikis and Loss of Adhesion

Written By

M.E. Hernández-Caballero

Submitted: 29 November 2011 Published: 23 January 2013

DOI: 10.5772/55399

From the Edited Volume

Carcinogenesis

Edited by Kathryn Tonissen

Chapter metrics overview

3,692 Chapter Downloads

View Full Metrics

1. Introduction

During the process of tumorigenesis, some cells separate themselves from the tumor to invade distant tissues. Cellular migration in this process is similar to what occurs during embryonic development and wound healing. Unlike these two healthy processes, which involve the creation of a structure or the healing of tissue, metastasis results in the formation of a cellular mass that, if not eliminated, leads to the death of the organism. The process of metastasis includes the invasion of the basal membrane and nearby tissue by tumoral cells and the intravasation towards the blood vessels or infiltration of the lymphatic vessels. This is followed by the mechanisms of survival of tumor cells in these vessels and their extravasation to different tissues of the organism, where they may be able to proliferate. This pathogenic process requires a precise coordination of various signaling pathways that allows the tumor cells to move across the cell membrane, remodel the matrix, transport themselves by circulation [1] and create the appropriate conditions, at a distance, for establishing themselves in a different organ (Figure 1).

Although exploring this complex process of motility and invasion by tumoral cells is fundamental for deepening the understanding of metastasis [2], much remains a mystery despite the enormous amount of research contributions. Both in vitro and in vivo, the time required for analyzing the evolution of this pathogenesis is excessive. Today the best way to make functional evaluations of the genetic changes that take place in humans with metastasis is with animal models. Although time consuming, a complete follow up can be carried out of the entire process, from the moment of the appearance of a primary tumor, to the strategies used by cancerous cells to escape from the controls of adhesion, their interaction with endothelial cells during their migration, and the establishment of a secondary tumor through the preparation of a new microenvironment favorable to tumor growth in the affected organ.

Figure 1.

Separation of tumoral cells from a primary tumor and its migration to reach a blood or lymphatic vessel for dissemination to a secondary site is a very complex process that includes changes in the expression of multiple genes, which are genes involved in cell adhesion, survival, chemoattraction, growth factors, miRNAs. In the figure you can see some of the genes involved throughout this process.

Advertisement

2. Primary tumor migration

The majority of deaths from cancer are due to metastasis. Nevertheless, in any given moment only a small proportion of tumoral cells acquire the capacity of invasion and dissemination [3]. This capacity is favored by the activation of signaling pathways that trigger the metastatic cascade, which results from the continuous exposure to the development of the primary tumor, to growth factors, to angiogenesis and to accumulated genetic changes [4]. Since the probability that tumoral cells that travel to new sites give rise to a tumor is very small [5], the process of tumorigenesis is actually quite lengthy. Yachida et al [6], after analyzing autopsies of victims of metastatic pancreatic cancer, proposed a quantitative model of tumor development and metastasis. They consider that at least a decade must pass between the occurrence of the initial mutation and the birth of the parent cancerous cell that eventually results in a tumor. Another five years or more are required for the acquisition of a metastatic capacity. After this latter event, the process takes place very quickly, with the life expectancy of two years for the affected patient. Hence, the challenge is to detect a tumor after the onset of the parent cancerous cell but before metastasis.

Some years ago, Engel et al [7] found that the period from the onset of metastasis to the diagnosis of breast cancer was approximately 6 years. For the purpose of establishing such a diagnosis, two types of tumoral cells can be detected: those circulating in the blood and those disseminated in lymphatic nodes and bone marrow. Such tumor cells can persist for years, awaiting the adequate conditions in some organ to be able to establish themselves in a secondary tumor. In the event of the existence of both types of tumor cells in a patient, the likelihood of tumor development is quite high [8].

There are two proposed models for explaining metastasis. Firstly, there is the model of linear progression, which considers that tumoral cells pass through multiple successive rounds of mutations, resulting in the selection of the most apt for proliferation in a relatively autonomous manner, followed by the transport of these clones to a new site. Secondly, the model of parallel progression holds that tumor cells separate themselves from the primary tumor before the acquisition of the malignant phenotype, and that these cells then undergo a somatic progression and metastatic growth at a distant site [9, 10].

Detailed analysis of diverse data, including that from animal and computational models, suggests that dissemination is not a lengthy process, at least in breast cancer, prostate cancer or cancer of the esophagus [11,12]. Thus at least for these cancers, the model of parallel progression is best supported to explain metastasis from the primary tumor. Through the use of comparative genomic hybridization, Baudis [13] analyzed 5918 malignant epithelial neoplasias and observed typical imbalances: whereas there were recurrent findings of increases in 8q2, 20q, 1q, 3q, 5p, 7q and 17q, similar patterns of losses were seen in 3p, 4q, 13q, 17p and 18q, among others. These genetic similarities between primary and metastatic tumors tend to indicate a convergent evolution more than the result of selection starting from a clone [5]. Nowadays it is well known that tumoral cells abandon the primary tumor before cancer is diagnosed. The possibility that these cells develop a tumor in a distinct organ depends largely on the characteristics of the primary tumor.

2.1. Cell invasion

Tumor cells must acquire certain characteristics for cell migration to occur, such as polarity and disassociation from their point of origin. Afterwards, these cells must undergo cycles of extension and contraction, and at the same time they adhere to and are released from the substrate [14] to move from one site to another. During this process their form is radically modified.

The ability of cells to migrate, studied since 1863 after the discovery by Virchow, allowed them to carry out various processes including embryogenesis, angiogenesis and wound healing to immune response [15], Cell migration can be divided into stages according to the changes observed in the morphology of the cell: polarization, protrusion, adhesion, translocation of the cell body, and retraction of the rear portion. The physiology of cell migration is diverse, depending on the type of cell involved [16]. For instance, fibroblasts and melanocytes generally migrate mesenchymally, as individual cells that are highly adhesive and require proteolytic remodeling of the matrix [17,18.]. These cells form specialized protrusions of the membrane, among which are lamellipodia, which are actin projections of the cytoskeleton formed in the extreme front part of the mobile cell, and invadopodia (invasive pseudopods), which are proteolytically active protrusions of the plasmatic membrane that are responsible for the focal degradation of the components of the extracellular matrix (ECM). The latter type, characteristic of highly invasive tumor cells, contains sites of matrix metalloproteinases (MMPs) [19]. Wolf et al [20] observed that tumor cells do not always require MMPs, since they can escape through openings in the ECM by means of a great contractile force. Also among specialized protrusions of the membrane of metastatic fibroblasts and melanocytes are philopodia and podosomes. The former protrusions are in the shape of thin bundles of actin, and are in charge of probing the environment in search of signals [21]. The latter are functionally the same as invadopodia, forming in macrophages and osteoclasts associated with a tumor, and aiding in the process of invasion. The capacity of malignant cells to migrate in a directional manner is due to the presence of receptors on their surface that allows them to follow the gradient of chemokines [22]. Among the most common type of chemokines are CXCR4 and CCR7, which have been found in diverse types of cancer and are attracted by CXCL12 and CCL21, respectively [23].

2.2. Mechanisms of invasion

Yilmaz et al [24] suggest classifying the invasion by cancerous cells as individual or collective. The use of one or another type of mobility depends on the type of malignant tumor and the surrounding tissue, defined by distinct patterns in the activity of extracellular proteases, matrix-cell adhesion mediated by integrins, cell-cell adhesion mediated by cadherins, cellular polarity and cytoskeletal arrangements [25]. Epithelial cells, which are the most common source of diverse cancers, can exhibit multiple phenotypes of migration. They commonly have a stationary behavior and form layers of cells interconnected with strong bonds. They are capable of moving individually, as a layer, or as a tubular structure. The latter type of structure is found during embryonic development [26]. Among epithelial tumoral cells, all these three types can be found for any particular tissue, suggesting that these pathogenic cells have the capacity to change from individual to collective migration, according to the environment that they face.

2.3. Individual cell migration

The movement of cells inside of living organisms is highly complex and strictly coordinated. For instance, hematopoietic cells exhibit a highly individualized ameboid movement, with little adhesion and without causing remodeling of the matrix. Tumor cells are capable of taking advantage of the strategies of healthy cells, moving in the same individual manner for example as a leukocyte or fibroblast. Individual migration of cells can have some variants: ameboid or mesenchymal [27, 28], solitary or in single file. When cells are transported to a new location, their form is modified. They often, but not always, adopt a bottle form. The constriction of their apexes can have two functions: (i) the cells can transport much of their intracellular content and begin the movement out of the epithelium, and (ii) they can reduce the amount of non-adhesive apex membrane, since by passing through the epithelial layer to arrive to the new location the apical point breaks. Additionally, this form allows for passage through the epithelium layer with a reduced space left behind. The apical constriction is driven by contractions based on actinomiosine, while the apical membrane is reduced by endocytosis. The cells must carry out a process of de-epithelialization, which means that they lose an essential property of epithelial cells— contact with neighboring cells.

Apart from the eventual loss of the adherence of the apical point, the adjoining surfaces forming the bottle neck are also broken up [29]. Due to employment of intravital multiphoton imaging, it has been possible to observe tumor cells in movement, and therefore to see breast cancer cells utilizing ameboid movement to transport themselves rapidly (~4µm/min) [30]. During transport these cells acquire a polarized phenotype, bearing Ca2+ in front of and behind the cell, which allows for the retraction of the rear portion [31]. This process of retraction is supported by the contraction of miosine II and by the disassembly of focal adhesion at the rear of the cell, due to the breakage mediated by calpain of the proteins of focal bonds, including integrins, talin, vinculin and focal adhesion kinase (FAK) [32]. Focal adhesion is composed of a structural point and adhesion signaling between the ECM and the cytoskeleton. The velocity of the formation and disassembly of the focal adhesions is what determines the efficiency of cellular migration [33]. In tumor cells the loss of E-cadherin facilitates the formation of focal adhesion and better communication with the ECM. The small STPases (Rho, Rac, CDC42) are among the key molecules for initiating remodeling of the cytoskeleton. The remodeling of actin is mediated by actin-related proteins (such as ARP2/3) [34]. The other type of individual migration, mesenchymal migration, can be used by cells that undergo a transition from the epithelial to mesenchymal cell type, the latter of which is present in 10-40% of carcinomas [35]. This type of migration requires proteolysis of ECM proteins so that the cells can move through the matrix-filled spaces of the tumor [19].

2.4. Collective cell invasion

Targeted multicellular migration can be of two types: (i) collective cellular migration, in which the cells undergoing transport maintain close contact with one another, and (ii) streaming, in which these cells are not always in direct contact [22, 23]. This latter type is carried out by cells during the stage of gastrulation in embryonic development and in wound healing. Collective cell migration, where the cells are bonded together in layers, is more common than individual migration under normal physiological conditions [36]. The best understood collective cell migration is that of wound healing. Neurons and smooth muscle cells also move collectively, as a cohesive group and in a coordinated and integrated manner. The cells involved in this movement retain much of their epithelial characteristics, such as the cell to cell adhesion, averting the need for an epithelial-mesenchymal transition that leads to a modification of cellular shape [37]. In all types of collective migration, the foremost cells of the migratory group actively participate in chemotaxis and degradation of the matrix in order to make way for passage. These leading cells are linked to the cells behind them, dragging them through the remodeled matrix [36].

In tumor cell collective migration, the leading cell can be a tumor cell with proteolytic activity or a stromal cell of the tumoral microenvironment [38]. Among the most relevant proteins for collective cell migration are the β1 and β3 integrins grouped in the ruffling edges of the cell group in movement. These proteins are responsible for providing the adhesion and dynamic strength necessary for transporting the cell group. The leading cells have the greatest proteolytic capacity in relation to the ECM, and it is their production of the metalloproteinases MT1-MMP and MMP-2 that enable proteolysis, as these proteins cut collagen fibers and form a pathway for collective migration and expansion of the migratory group [39, 40]. In squamous carcinomas of the larynx, lungs, esophagus, cervix and skin, among other tissues, collective invasion has been associated with the presence of podoplanin and CDC42 proteins [41-43].

Advertisement

3. The epithelial to mesenchymal transition (EMT)

Regarding the acquisition of mobility, a fundamental process in the transformation to a malignant cell, the best understood phenomenon is the epithelial-mesenchymal transition. The epithelial layer is formed by cells with apical-basal polarity that maintain the laminar structure intact through various types of adhesion, thus forming the barrier necessary for the good functioning of the epithelial layers. This barrier, known as the apical joint complex, is formed and maintained by cell to cell contact at the cell surfaces that surround the apical-lateral dominion. This complex includes the adhesive joints (AJ), which promote the integrity of the tissue by maintaining a strong bond between the cells [44] and have cadherins as their principal component [45]. The resulting tight joints (TJ) form a physical barrier to the movement of ions, macromolecules, immune cells and pathogens between the epithelial cells [46] and desmosomes. The AJ as well as the TJ are closely related with the actin cytoskeleton and are functionally regulated by filaments of the circumference of actomyosin [47]. Their dynamic interaction is important during the morphogenesis of epithelial tissues in embryonic development, in tissue repair [48], and in the maintenance of an effective epithelial barrier in an adult [49].

During early embryonic development, there is a process of essential differentiation resulting in the establishment of the germinal layers in the epithelial-mesenchymal transition (EMT) [29]. The EMT, discovered by Frank Lillie in 1908, is an important mechanism for the reorganization of the germinal layers [50]. This process, whose essential characteristics are the interruption of intercellular contacts and the acquisition of a fibroblast-type conjugated morphology, converts well-organized epithelial cells into isolated cells with a mesenchymal morphology that are capable of transporting themselves [51] to become established in new tissues, thus allowing for inductive interactions during embryogenesis. Currently, it is thought that three subtypes of EMT exist: EMT type 1 is related to embryonic development, EMT type 2 to wound healing, and EMT type 3 to tumor cells [52]. As Thiery [53] points out, it took a long time for EMT to be recognized as a mechanism of tumor progression due to the difficulty of detecting the process in patients. Today, it is known that tumor cells utilize the EMT as a key element in the invasive process, a phenomenon very similar to embryonic EMT. There are diverse pathways in common between embryonic and tumoral EMT, including the stimulation by TGF-β that induces phosphorylation of β-catenin, which in turn activates transcription factors such as Snail [54], as well as diverse growth factors and proteins. This is accompanied by a loss of diverse epithelial proteins such as E-Cadherin, α and β-catenins, Claudin, ocludin and citokeratins, and at the same time an overexpression of mesenchymal proteins such as vimentin, fibronectin, metalloproteinases, actin and integrins αv and β1 [55, 56]. E-cadherin is a key protein because it facilitates the adhesion of epithelial cells and desmosomes [57], which in turn prevents cellular mobility and metastatic dissemination.

The cadherin switch is essential to increase mobility, but is not always necessary for the morphological changes that accompany the EMT [58]. The expression of cadherins can be inactivated by somatic mutations, hypermethylation and desacetylation of histones or transcriptional repression. Some researchers have considered that the expression of N-cadherin is more important for metastasis than that of E-cadherin [59], based on the fact that the former is a mesenchymal protein marker found overexpressed in cells with the EMT. Camand et al [60] found that a decrease in the N-cadherin levels, through regulation of focal adhesion formation, promotes a more rapid migration in healthy and tumoral glia cells. In the latter cells, this contributes to the invasive capacity of astrocyte tumors.

Integrins, a determining factor in the process of cellular adhesion, can also induce EMT. Recently, Gupta et al [61] proposed that integrin α9β1 is implicated in EMT by accelerating lung tumor cell migration, mediated by scr, in a TGFβ-independent manner. To date, a wide variety of proteins and factors have been discovered that regulate EMT. Indeed, it is thought that hundreds of genes significantly modify their expression during EMT [52]. One example is Twist, a potent inducer of EMT that was originally identified as an inducer of the mesoderm in Drosophila [62]. Its expression in epithelial cells causes the loss of cell to cell adhesion mediated by E-cadherin, which leads to cell decomposition. It also activates BMI1 polycomb proteins, which induce stem-like properties and thus annul the p53- and Rb-dependent pathways that would otherwise allow the cells to enter in premature senescence induced by oncogenes. Twist also suppresses the production of let-7i, which in turn allows cells to acquire mobility in the last stage of EMT [63, 64]. The overexpression and activation of the signaling pathways of HER-2/Neu and TGFβ activate EMT in tumoral cells of breast cancer.

EMT is also regulated by alternative splicing, such as that of ENAH, a regulating protein of the cytoskeleton of actin. The transcript present in epithelial cells contains a small exon 11a, which is absent in mesenchymal cell lines and during the EMT [65]. RBFOX2 is a splicing factor which together with ESRP1 and ESRP2, proteins for binding with RNA that are specific to the epithelium, promote splicing of transcripts that are important in EMT [66]. The loss of ESRPs in epithelial cells induces morphological changes similar to EMT [67]. On the other hand, Shapiro et al [68] found a partial induction of the splicing program in mesenchymal cells by means of the ectopic expression of ESRP1 or by elimination of RBFOX2 in immortalized epithelial mammary cells. Mesenchymal isoform IIIc of FGFR2, isoform 3 of catenin p120, and isoform CD44s of CD44 are all expressed in mesenchymal cells and in tumoral cells with the EMT, as reviewed in [69].

Just as the process of EMT resembles that observed during embryonic development, it seems that both pathways are regulated by an intricate interaction of transcriptional and post-transcriptional programs, as was discovered by observing that the ribonucleoprotein, heterogeneous nuclear E1 (hnRNP E1) is a selective transductional regulator of transcripts of EMT induced by TGFβ. Due to its capacity to bind to RNA, hnRNP E1 adheres to 3'-UTR BAT structural elements present in the transcripts of mRNA necessary for EMT, and upon doing so silences these elements. This silencing, mediated by hnRNP E1, occurs during the stage of elongation when hnRNP E1 impedes the release of the elongation factor eukaryote 1 A1 (eEF1A1) of the ribosomal site A. The phosphorylation of hnRNP by Akt2, mediated by TGFβ, restores the transduction of white transcripts of EMT. The attenuation of the expression of hnRNP E1 in non-invasive breast cancer cell lines induces not only EMT but also metastasis [70,71]. Furthermore, the steroid hormones such as androgens have also been observed to be implicated in the maintenance of cellular characteristics, evidenced by the fact that Sun et al [72] found that the absence of androgens in prostate tumors is associated with a worse prognosis and acquisition of cell characteristics typical of mesenchymal or stem cells.

With triple negative breast cancer (ER-, PR- HET2-), there is an overexpression of N-cadherin and EMT, leading to a worse prognosis. Regulation by miRNAs is not an exception in EMT, particularly the miRNAs of the miR-200 (miR-200a, miR-200b, miR-200c, miR-141 and miR-429) and miR-205 family. These miRNAs participate in the regulation of EMT induced by TGFβ by mediating the production of ZEB1 and ZEB2 [73,74]. Recently miR-29b was added to the list of miRNAs that regulate EMT, due to the fact that an increase in miR-29b in PC3 prostate cancer cells inhibits the capacity of these tumoral cells to invade and colonize [75]. In addition to TGFβ, other growth factors such as EGF, HGF, PDGF, FGF2, TNFα and IGF can induce EMT by activating the expression of transcriptional repressors of E-cadherin or by activating the transduction signaling pathways, such as MAPK, PI3K, Wnt/β-catenin, NF-κB and Notch [69 and references there].

3.1. EMT reversion

After tumoral cells are disseminated, they must reactivate their epithelial properties by means of the reversion of the process of EMT, known as the mesenchymal-epithelial transition (MET). In the metastatic process, just as EMT is essential for the initial transformation of cells from being part of the epithelial tissue to being released from intercellular connectedness and ready for transport, MET is critical for the later stages of metastasis. Possibly this latter change is triggered by the local environment after extravasation towards the parenchyma of the invaded organs [76]. Although EMT has been extensively studied, there is much yet to be discovered about the molecular mechanisms that regulate MET. One of the keys to the process of MET is the re-expression of E-cadherin, which enables tumor cells to interact with the tissue of the recently colonized organ [77]. Leontovich et al [78] demonstrated that the constitutive activation of Raf-1 induces the overexpression of HER-2/Neu, leading in turn to the development of metastasis in xenografts of MCF7 ERα+ cells. This metastasis was linked to the activation of the MET pathway, and curiously it was characterized by a reduction in the expression of genes implicated in EMT, such as TGFB2, TWIST and FOXC1. On the other hand, Phino et al [79] found that the levels of glycosylation of E-cadherin regulated by Mgat3/GnT-III were diminished in EMT, and that they were recovered when the cells once again acquired an epithelial phenotype. Other genes identified in MET are WT1, BMB7, WNT4 and the protein formina IV of the cytoskeleton [53 and references there]. Diverse miRNAs have been found to be deregulated in tumors, particularly metastamir, which is implicated in metastasis [80]. Wang & Wang [81] summarized the types of metastamir that are deregulated in breast cancer cells: miR-9, -10b, -21, -29a, -31, -103/107, -126, -335, -210, and -373. All of these are involved in migration, aggressiveness and the worst prognoses. An increase in the levels of miR-200 induces MET by reducing motility and aggressiveness [82]. Likewise, the expression of miR124 modifies the morphology and capacity of metastasis of MDA-MB-231 cells, as well as by decreasing the levels of vimentina, a mesenchymal marker [83]. The regulation of diverse transcription factors, receptors for growth factors (including FGFR2b, FGFR2c, EGFR and HER2), and the activation of Akt are other elements in the reversion of MET.

3.2. Collective to ameboid transition and the mesenchymal to ameboid transition

Through the process of metastasis, cells can undergo changes in the way in which they transport themselves. In explants of melanoma, it has been observed that the inhibition of the integrin β1 provokes the separation of groups of cells, which thus become individual ameboid cells. These cells acquire the capacity to migrate, independently of proteases and/or integrins, similar to that observed in ameboid movement. Hence, the transition from collective invasion to ameboid movement of individual cells is an interchangeable and bidirectional process that depends on environmental factors, making evident the great plasticity that tumoral cells can have [84]. The mesenchymal-ameboid transition can serve as a compensatory mechanism of migration after the inhibition of pericellular proteolysis. This transition provokes a change from a cellular form similar to a fibroblast to a round or elliptical shape, and at the same time spurs a change in the cytoskeletal organization as well as the distribution of integrins [85].

Advertisement

4. The tumor microenvironment

During the process of migration, cancerous cells pass through different microenvironments, including the stroma, the endothelium of blood vessels, the lymphatic vessels and the tissue of the organ where the secondary tumor forms [86]. In each of these sites, the microenvironment exerts a strict control over the behavior of tumor cells [87].

Cells can cross the basal membrane (BM) during embryonic development and immune vigilance, either as part of the embryonic process or more routine morphological processes such as leukocyte activity [88]. The BM is a dense layer of the extracellular matrix that is highly reticular and rich in laminina, collagen type IV, perlecan, nidogen-1 and nidogen-2. Its structural function is to delimit epithelial and endothelial tissue, and to give polarity to cells [89, 90]. After the loss of intercellular adhesion, cells acquire a mobile phenotype and produce metalloproteinases that allow them to digest the BM. Hence, the BM is the first barrier that tumor cells must cross [2]. As a tumor invades an organ, there is a dynamic interaction between invasive cells, the invaded tissue and the BM that separate the two. Although it has not been easy to study this process in vitro, the transmigration of leukocytes has proved to be a very useful model, allowing for the identification of regions of the perivascular BM that have less resistance to invasion. These regions contain reduced levels of laminin and collagen IV [91]. It is not yet clear how carcinoma cells initiate the invasive process after abandoning the primary tumor, but there is evidence that points to the participation of invadopodium in the proteolysis of vascular BM.

4.1. Interactions with the extracellular matrix

A critical component of the cellular microenvironment is the extracellular matrix (ECM), a biopolymer complex that provides healthy and tumor cells with biophysical and biochemical signals that influence their function and survival [92]. Cells are capable of sensing and responding to changes in the conformation of the ECM through a process of mutual feedback that involves cellular contractile mechanisms. Each cell type responds to the rigidity that is characteristic of its host [93]. Integrins are crucial participants in the communication of cells with the ECM, because they act as a conduit between extracellular ligands and the cytoskeleton. Therefore they have the capacity of responding to the rigidity of the external substrate through a counter-response exerted by the actomiosin network. They act as mechanosensors that undergo conformational changes in response to mechanical forces, resulting in increased adhesion between the cells and the ECM, the formation of focal adhesions, and the dispersion of the cells [94]. The extracellular signals mediated by integrins are transduced internally through focal adhesion components [95]. Tumor cells have the capability of ignoring the conformational changes that take place in the ECM, and this capability is associated with the activation of integrin β1 and the phosphorylation of FAK in a manner dependent on fibronectin and the overexpression of αvβ3 [96]. Lysyl oxidase (LOX), a copper-dependent amine oxidase that catalyzes the crosslinking of collagen, elastin and fibrin in the ECM, can modulate metastasis by facilitating extravasation or by conditioning the metastatic niche, the latter done by increasing the rigidity of the ECM and fibrillar collagen [97]. As can be seen, although the ECM is considered a barrier for the dissemination of tumor cells, its remodeling by tumor cells actually converts it into a promoter of the same process. The release of proteins from the ECM by tumor MMPs can create a promigratory stimulus, in part by increasing the rigidity of the tissue, which in turn augments the motility of tumor cells [98].

4.2. The cancer-associated fibroblast and the tumor associated macrophages and immune cells

Diverse signals emanating from tumor cells allow them to recruit and activate host cells, particularly monocytes, fibroblasts and mesenchymal stem cells, all of which are abundant in the microenvironment of a tumor. Among these cell types, fibroblasts are the most abundant and important in the interaction with the tumor. The carcinoma associated fibroblasts (CAFs) are miofibroblasts with contractile properties and a staining capacity for alpha-smooth muscle actin (α-SMA). Several studies have reported that they can potentiate tumorigenesis by expressing proteins that favor migration or by altering the stromal environment in a way that facilitates colonization. Brentnall et al [99] point out that the overexpression of paladin by the activation of k-ras can transform normal fibroblasts into miofibroblasts in 5 days. As a result, the expression of paladin alters the morphology and behavior of cells, leading to an increased cellular migration, the degradation of the ECM by the formation of invadopodia, and the creation of tunnels through which cancerous cells can transport themselves. Malanchi et al [100] found that to facilitate the process of colonization, infiltrating tumor cells need to induce the expression of stromal periostina (POSTN) in the target organ for a secondary tumor. POSTN is a component of the ECM that is expressed by fibroblasts, both in healthy tissue as well as in the stroma of a primary tumor. Møller et al [101] reported that fibulin 5, whose expression is downregulated by factors released by fibroblasts in tumors, is capable of suppressing metastatic colonization of the lungs and liver by inhibiting the production of metalloproteinase 9 and by reducing the invasive behavior of fibroblasts. Hence, tumor cells avoid the expression of this protein.

Epithelial cells produce multiple factors that recruit various cells able to facilitate the infiltration of tumors, including tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), lymphocytes, mesenchymal stem cells, endothelial cells and the aforementioned CAFs. In the case of macrophages, experimental evidence exists supporting the idea that they can differentiate into pro-inflammatory macrophages (M1) or anti-inflammatory macrophages (M2), depending on the stimulus [102,103]. TAMs are similar to M2 macrophages, promoting tumor progression by their incapacity to induce T cells, their increased expression of manose and scavenger receptors, as well as their release of pro-tumorigenic factors (e.g., TGF-β1, IL-10 and MMPs) and pro-angiogenic factors [104]. As they generally accumulate in hypoxic zones, the pro-angiogenic program of TAMs is triggered, resulting in the secretion of VEGF, IL-1b, TNF-a, angiogenin and semaphorin 4D. By secreting urokinasetype plasminogen activator (uPA) and its receptor, uPAR, TAMs can participate in the degradation of the ECM [105].

The non-neoplasic cells present in the stroma of the tumor produce a series of chemokines and growth factors that form a complex network of communication within the tumoral microenvironment. One factor produced by the CAFs is CXCL12, which promotes the migration of tumor cells towards blood vessels [106]. TAMs co-migrate with tumor cells in a paracrine-dependent manner. They produce EGF, which increases the migration of breast cancer cells that express EGFRs. In response, these tumor cells secrete CSF1, thereby attracting the TAMs that express CSF1Rs [107]. Campbell et al [108] found that the TAMS positive to CD68 and PCNA are associated with a worse prognosis for breast cancer patients.

Macrophages are capable of regulating other stromal cells and of exerting influence through them. For instance, the loss of signaling by TGF-β in mammary fibroblasts produces an increase in the secretion of CCL2, which in turn results in a progression of 4T1 tumors, either through direct action on cancerous cells or through the recruitment of macrophages [109]. The infiltration of lymphocytes is associated with metastasis and an increased expression of the activator of the NF-κB (RANK) receptor and of its ligand (RANKL). In particular, T regulatory cells (Treg) infiltrated in a breast cancer tumor stimulate metastasis in breast cancer through the signaling by RANKL-RANK to IKK-α, causing the suppression of the inhibitor of the antimetastatic serine proteinase and maspin, and favoring the survival of circulating tumor cells [110]. It is possible that CAFs express CCL5, thus attracting Treg cells that express CCR1 and produce RANKL. Treg cells secrete IL-4 and IL-13, which in this case would result in the activation of TAMs and consequently the promotion metastasis in RANK-positive tumors [111]. Neutrophils, on the other hand, participate in the preparation of the tumoral microenvironment. They produce enzymes that remodel the ECM, and also produce ROS, which in turn activates NF-κB and in this way allows tumor cells to attenuate their apoptotic response and increase their mutation rate [112].

Advertisement

5. Survival of tumoral cells and modulation of cell survival

The metastatic process has various rate-limiting steps, and consequently only a minority of tumor cells is able to reach distant sites [86]. It has been suggested that approximately 0.01% of circulating tumor cells can manage to produce a metastasis [113]. Talin 1 is one of the proteins whose altered expression allows for the survival of tumor cells. It can recruit focal adhesion proteins ILK, FAK and Src through its interaction with integrin β, resulting in the promotion of the survival, invasion and angiogenesis of tumor cells. Its overexpression allows prostate cancer tumor cells to activate survival signals and resist death by anoikis [114]. In this sense, the PI3K/Akt and Wnt/β-catenin signaling pathways, along with mutations in p53 and other genes, play an important role in the avoidance of death by metastatic cells.

The adhesion of tumor cells to the ECM is not sufficient for inducing a survival signal. The changes in the cytoskeleton associated with adhesion signaling are critical for tumor cell survival. Halder & Johnson [115] indicate that the newly established Hippo suppressor pathway (which regulates the size of organs by maintaining a balance between proliferation and apoptosis in physiological conditions) might be involved in tumor proliferation. The Hippo pathway phosphorylates and inhibits the transcriptional coactivator Yes-associated protein (YAP), a key oncogene for the regulation of the size of organs. This inactivation is triggered by the reorganization of the cytoskeleton. Once the cell has separated, the Hippo kinases Lats 1/2 are activated, causing the inhibition of YAP, and therefore resulting in anoikis. It has been found that with metastatic prostate cancer, the levels of expression of Lats 1/2 are significantly downregulated [116], whereas, Lamar et al [117] demonstrated that increased YAP activity promotes metastasis and tumor growth at both the primary site and the metastatic site.

5.1. Integrins in cell survival and apoptosis (anoikis)

A loss of balance between cell division and cell death is common in cancer, due to a decrease in apoptotic cell death that leads to the progression of a tumor. There are various ways by which a malignant tumor cell averts or resists apoptosis.

The distinctive morphological characteristic of apoptosis is the condensation of chromatin and the posterior fragmentation of the nucleus, accompanied by a reduction of cell volume, a retraction of pseudopods, and the formation of vacuoles, all leading to a loss of integrity of the cell membrane [110]. The biochemical changes that occur during apoptosis are: (i) the activation of caspases, (ii) the rupture of DNA and proteins, and (iii) changes in the membrane related to its recognition by phagocytes [111].

Tumor cells can avert apoptosis by altering the balance of pro-and anti-apoptotic proteins, decreasing the function of caspases, and altering the signaling of receptors related to cell death [112]. A cancerous cell in an ectopic site, whether near a primary tumor or in route to a secondary organ, can employ various mechanisms to avert the process of apoptosis known as anoikis [113]. Through anoikis, a healthy cell in an inappropriate location activates its programmed death by separating itself from its neighbors and its microenvironment, and thus is eliminated. Anoikis is therefore a barrier against the formation and survival of potentially oncogenic clones. Only non-adherent cells, such as leukocytes and mature hematopoietic cells, are protected from anoikis [114].

The principal effector mechanisms of anoikis are autophagy and apoptosis [115]. A critical step in the series of changes through which a tumor cell passes to avoid apoptosis is a change in the expression of integrins, which are a family of receptors that receive signals from the ECM. This change is based on genetic and epigenetic alterations that can only occur in the microenvironment of a tumor [116], allowing tumor cells to ignore signals by the ECM and act as if they were in the appropriate microenvironment. Besides this extrinsic pathway, tumor cells can also avoid apoptosis by damaging the mitochondria, an intrinsic pathway allowing these cells to hyperactivate mechanisms of survival and proliferation [117]. Another important factor in anoikis is the integrity of the cytoskeleton [118], since many pro-apoptotic proteins, including BIM and BMF, co-locate with the cytoskeleton.

In vertebrates, integrins are a family of receptors composed of 18 subunits α and 8 subunits β, whose combination gives rise to 24 types of these receptors [119]. Changes in the integrins that act as receptors for the ECM are essential for the avoidance of anoikis. The normal functioning of these receptors not only provides a physical bond with the ECM, but also establishes a platform that depends on signals from adhesion molecules, including adaptor proteins and kinases [120, 121]. For instance, the integrins α1β1, α2β1, α3β1, α5β1, α6β1, α6β4 and αvβ3 have a profound impact on cell survival [122].

A particularly important structure in this sense is hemidesmosome, in which there are complexes of integrin-talin-paxilin-actin that receive signals from the ECM and transduce these signals through various proteins such as focal adhesion kinase (FAK) in order to avoid anoikis. FAK, the kinase linked to the integrin ILK, tyrosine kinase Src, PI3K, the extracellular signal-related kinase (ERK) and the adaptor protein Shc are key to the transduction of signals mediated by integrins for protection against anoikis [123]. The FAK binds to the cytoplasmic tails of the integrins and is autophosphorylated at the Y397 residue in order to transmit a survival signal [124]. After the adhesion of integrins with the appropriate proteins of the ECM, FAK and ILK recruit and activate PI3K/Akt, ERK and the Jun kinase (JNK) pathway [125].

The separation of these cells, which move towards the vascular lymphatic space, as well as their implantation in a site with an unknown ECM can cause the separation of the α and β subunits of the heterodimeric receptors of integrin, which in turn leads to the deactivation of FAK, of the family of tyrosine kinases Src, and of ILK. Whereas this attenuates pro-survival pathways, including the mitogen-activated protein kinase/ERK, Akt/phosphatase, tensin homologue (PTEN), and the nuclear factor κB (NF-κB) [126], it at the same time stimulates other mechanisms of survival. The latter mechanisms include the activation of PKB/Akt, which can inhibit anoikis at multiple levels. This inhibition takes place by the inactivation of caspase 9, by a scavenging process that is dependent on the phosphorylation of the pro-apoptotic Bad protein by the 14-3-3 protein, by the activation of NF-κB, and by the inhibition of transcription factors Fork-head [127-130].

It has been seen that the response to anoikis can be recovered by silencing FAK with siRNAs in pancreatic cancer cells [131]. Recently it was found that the inhibition of the proteins of the subfamily of Rho and the expression of Akt in the B16F10 cell line inactivates the FAK pathway and induces anoikis in resistant cells [132]. Whereas healthy epithelial cells express the receptor for collagen α2β1 and receptors for laminin α3β1 and α6β1, hyperproliferative cells overexpress integrins αvβ5 and αvβ6 and squamous carcinoma cells overexpress αvβ6. The latter integrin is required for the adhesion of melanoma cells to dermic collagen and for survival in the environment of the new tissue [133-137]. Goldstein et al [138] found that the substitution of valine by glutamic acid at aminoacid 600 of B-RAF (B-RAFV600E), which is found in 66% of melanomas, is an event that leads to resistance to anoikis by continuous activation of MEK/ERK and the inhibition of BIM, a pro-apoptotic protein. Whereas in a healthy cell BIM levels are increased upon its separation from its tissue as a mechanism to activate its programmed death, this does not occur in tumor cells. Although the overexpression of integrin αvβ3 is associated with an increase in invasiveness, it has also been implicated in the induction of anoikis, which suggests that it has a dual role in tumorigenesis [139, 140].

ANGPTL4, a regulator of the metabolism of lipids, leads to the activation of Rac1 and FAK by binding to integrins β1 and β5. This phenomenon protects against anoikis through the activation of Src, Akt/PKB and ERK. Hence, the binding of ANGPTL4 with integrins is capable of deceiving the mechanism of programmed death by generating a false impression of anchorage [141]. The phosphorylation of ERK2 by FAK, though, leads to the phosphorylation of BIM, marking the latter for ubiquitination and degradation. Another critical protein for resisting anoikis is the α5 integrin, which as shown by Shen et al [142], has its expression in gastric cancer regulated by the S100A4 protein. S100A4 permits the survival of tumor cells and favors the overexpression of the α5 integrin.

By interacting with integrins, the microenvironment modulates their function in an important way. Marchan et al [143] found that β3 expressed in pancreatic cancer cells has important effects, including the promotion of migration in single-layer cell cultures, the induction of anoikis in vitro, and the suppression of tumor growth in vivo. At the same time that tumor cells avoid programmed death by anoikis, they acquire a mesenchymal phenotype that allows them to begin migration.

Advertisement

6. Transendothelial or lymphatic migration and extravasation

Tumor cells must go in and out of blood vessels by crossing endothelial layers as part of the metastatic process, and in order to do this they undergo dramatic changes in their shape, driven by a reorganization of their cytoskeleton. The most difficult part of these changes in malignant cells, which adopt elastic and viscous properties for this purpose, is the deformation of the nucleus in interphase, because it is 10 times more rigid than the cytoplasm [144].

Intravasation is the principal route for the dissemination of tumor cells coming from carcinomas. Once inside of a blood vessel, the survival of tumor cells is affected by various factors, including hemodynamic forces, immunological stress, and collisions with blood cells. Consequently, only a very small number of tumor cells manage to leave blood vessels [145]. The majority of tumor cells that enter the bloodstream are detained in the microvasculature of the first organ they pass through [146].

It has been demonstrated that in breast cancer tumors, intravasation depends on the paracrine loop between tumor cells and TAMs [100]. In one clinical study of patients with breast cancer, the half-life of tumor cells in the bloodstream was from 1 to 2.4 h [147]. Png et al [148] pointed out that endothelial cells play an important initial role in metastasis by providing signals that promote this process. These researchers identified molecules such as secretory IGFBP2, the transference protein PITPNC1, and the kinase MERK as necessary for mediating the recruitment of metastatic endothelial cells.

Brown & Ruoslahti [149] suggested that there are molecules that favor the capture of tumor cells in blood vessels. In the case of breast cancer cells, the expression of metadherin favors homing in the pulmonary vasculature. In the majority of tumors of epithelial origin, the first sign of metastasis is found in regional lymphatic nodes. Once inside the lymphatic vessels, tumor cells can produce local or regional metastasis, or can enter in a quiescent state, whose duration depends in great part on the immune system. Immune vigilance can induce quiescence in individual cells by detaining the cell cycle through signals mediated by cytokines [150, 151]. It is estimated that 80% of metastasis of solid tumors, such as in breast cancer and melanoma, are disseminated through the lymphatic system [151].

In the bloodstream a tumor cell can be detained or can succeed in passing through the wall of a blood vessel. If the tumor cell has a diameter greater than that of a given blood vessel, mechanical or occlusive detainment will occur. This mechanism of detention has been observed in mouse models of brain cancer metastasis [152]. On the other hand, if the tumor cell has a diameter less than that of the blood vessel, it must bond to the wall of this channel in order to be able to leave the bloodstream. There is evidence that tumor cells are associated with platelets, which can mask and protect these cells from being located by natural killer cells [153]. Platelets can also facilitate the accumulation of VEGF at the bond between a tumor cell and the endothelium tissue, favoring vascular hyperpermeability and consequently extravasation [154].

Advertisement

7. Colonization

During the progression of cancer, the colonization of distant organs by circulating tumor cells marks the difference between a possibly curable tumor and a systemic and generally incurable disease [155]. After an analysis of breast cancer tumors, Paget [156] concluded that the local microenvironment of some organs must be favorable for the dissemination of tumor cells or their progenitors. On the one hand, the microenvironment of the target organ is a determinant for the survival of tumor cells, and on the other hand the accumulation of these malignant cells in a given organ can create an adequate stromal microenvironment to allow metastasis [157].

In the preparation of an adequate niche, the primary tumor secretes VEGFA to mobilize progenitor hematopoietic cells from bone marrow (HPCs) toward the bloodstream, and from there to the site of metastasis. The same process occurs with fibronectin, metalloproteinases and growth factors, all of which can be synthesized by the primary tumor and released to the bloodstream, later to accumulate in the target organ. In this way, these molecules prepare a premetastatic microenvironment, to which the HPCs arrive and continue this process of adaptation for the purpose of their own survival.

Tumor cells and macrophages are recruited towards the premetastatic niche due to the chemokines synthesized by cells associated with cancer [158]. It has been shown that chemokines are chemotactic molecules important for colonization of a given organ [159, 160]. Wendel et al [161], by utilizing intravital observation techniques, were able to demonstrate that the receptor of chemokine CXCR4 plays an important role in the extravasation of breast cancer tumor cells towards their target organs.

The anatomy of some organs provides a barrier to metastasis. In the case of the lungs, endothelial capillaries are surrounded by a basal membrane [162]. In the blood-brain barrier, capillaries have tight junctions and astrocyte foot processes [163]. Capillaries in bone marrow and the liver, on the other hand, are fenestrated, making them susceptible to invasion [162].

Another important factor in the ability of a tumor to invade a distant organ is the type of primary tumor. Breast cancer tumors positive for estrogen receptors, as well as ocular prostate cancer tumors and melanoma tumors, can give metastatic signals decades after elimination of the primary tumor [7, 164-166]. This is due to the fact that tumor cells are disseminated from the primary tumor various years before detection of the primary tumor. Adenocarcinoma of pancreatic and lung cancer have malignant cells that rapidly acquire the capacity to infiltrate and colonize without requiring a process of quiescence to generate a macrometastasis [167, 168]. Furthermore, tumor cells in general can adopt two distinct states of quiescence while awaiting a favorable microenvironment. They can exist as individual cells (cellular quiescence) or as small indolent groups (tumor mass quiescence), in either case maintaining a balance between proliferation and death during the period of latency [169].

7.1. Most frequent organs for metastasis

Although no organ is totally protected from the development of metastasis, this process tends to take place more frequently in certain regions of the body, including the lymphatic nodes (lymphatic metastasis), lungs, liver, bone marrow, brain (haematogenic metastasis), peritoneum and pleura [145]. Each type of cancer is more prone to certain organs. When it occurs, metastasis exists in at least 2 organs, with an average of 5.6 metastases per patient.

With breast cancer, metastasis generally is a slow process that occurs in the lymph nodes, lungs, bone marrow, liver or brain. Only in 6-18% of breast cancer patients does metastasis takes place in the gastrointestinal tract, with the stomach and small intestine being the most frequent hosts. On the other hand, colorectal cancer rarely metastasizes to bone marrow [170]. Adrenal glands are the preferred site for metastasis of lung cancer tumors with small cells [171]. In North and South America, lung and breast cancer are the first and second source of metastasis to the brain. Generally the earliest metastasis takes place with lung cancer [172], with 80% of such events occurring in the cerebellar hemispheres [173]. However, the most frequent metastasis to the cerebellar hemispheres occurs in breast cancer patients [174]. This frequency can be explained in part by the production of osteoclast-activating factors, such as the parathyroid hormone related protein (PTHrP), IL-1, IL-6, IL-11 and GM-CSF by breast cancer tumor cells [175]. These osteoclasts release growth factors derived from bones, such as TGF-β and IGF-1 [176]. Contrarily, the metastasis of tumor cells from prostate cancer to bone marrow generally stimulates the formation of bone, regulated by the production of osteoblast-activating factors such as endothelin-1, BMPs and PDGF. In this way, the activated osteoblasts can maintain the proliferation and survival of the tumor cells [177].

Cyclooxygenase COX 2, EGFR, the ligand HBEGF and α2,6-sialyltransferase ST6GALNAC5 are mediators of the passage of cancerous cells through the blood-brain barrier. Nevertheless, only ST6GALNAC5 mediates metastasis of breast cancer tumor cells to the brain, facilitating the both the passage through the blood-brain barrier and the adhesion of these cells to brain endothelial cells [178]. A recent study [179] reported that the greatest number of metastasis to the brain occur in breast cancer patients of African origin. The least frequent teratogenic metastases are found in the kidneys, gonads, spleen, subcutaneous fat and especially in the walls of the gastrointestinal tract, uterus, heart and skeletal muscle [180].

The gradients of chemokines can explain the tropism of some types of cancer. It has been suggested that in organs with high levels of chemokine expression, these latter molecules can attract metastatic tumor cells that express the corresponding receptor. This model has been demonstrated in the metastasis of breast and prostate cancer tumors to bone marrow due to the presence of CXCR4-CXCL12. With solid and hematopoietic tumors, chemokines CCL21 and CCL19 are present in metastasis to lymphatic nodules with the corresponding receptor CCR7 [18].

Advertisement

8. Conclusion

The capacity of tumor cells to be transported to organs distant from the primary tumor is driven by two types of factors: the complex interactions between tumor cells and their surroundings, and the changes in such cells that allow them to adapt to their environment. Among the latter factors is the hallmark of tumor cells: their capacity to avoid apoptosis. At the onset of the metastatic process, the epithelial-mesenchymal transition is critical for a tumor cell to escape the controls of adhesion and be released for transport from the primary tumor. It must then be able to move across the cell membrane, remodel the extracellular matrix, transport itself by circulation, and establish itself in a distant organ. The mesenchymal-epithelial transition is critical for this last stage in which the tumor cell survives in a new organ. Although very few tumor cells are able to achieve this task, metastasis unfortunately is a common occurrence with cancer patients due to the continuous exposure to the development of the primary tumor, to growth factors, to angiogenesis and to accumulated genetic changes. The great complexity of the regulatory network that is implicated in the susceptibility of an organ to metastasis, as well as the capacity of tumor cells to dodge the immune response and influence the microenvironment of a given organ to make it favorable to metastasis, makes the study of metastasis for the purpose of drug design for cancer treatment a titanic task. The advantage is that there are now thousands of researchers and hundreds of thousands of reports in the field of metastasis. Furthermore, metastasis is a lengthy process, often taking a decade. Hence, the challenge is to detect a tumor after the onset of the parent cancerous cell but before metastasis.

References

  1. 1. FriedlPWolfK2003Tumour-cell invasion and migration: diversity and escape mechanisms.Nat Rev Cancer. 3362374
  2. 2. Bravo-corderoJ. JHodgsonLCondeelisJ2011Directed cell invasion and migration during metastasisCurr Opin Cell Biol. 2011 Dec 30.
  3. 3. WangWGoswamiSLapidusKWellsA. LWyckoffJ. BSahaiEet al2004Identification and testing of a gene expression signature of invasive carcinoma cells within primary mammary tumors.Cancer Res. 64858594
  4. 4. WirtzDKonstantopoulos K Searson PC (2011The physics of cancer: the role of physical interactions and mechanical forces in metástasis.Nat Rev Cancer. 11512522
  5. 5. KleinC. A2009Parallel progression of primary tumours and metastasesNat Rev Cancer. 9302312
  6. 6. YachidaSJonesSBozicIAntalTLearyRFuBet al2010Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 46711147
  7. 7. EngelJEckelRKerrJSchmidtMFürstenbergerGRichterRet al2003The process of metastasisation for breast cancer. Eur. J. Cancer. 3917941806
  8. 8. HynesR. O2011Metastatic Cells Will Take Any Help They Can GetCell. 20689690
  9. 9. KleinG1998Foulds’ dangerous idea revisited: the multistep development of tumors 40 years later. Adv. Cancer Res. 72123
  10. 10. FribergSMattsonS1997On the growth rates of human malignant tumors: implications for medical decision making.J. Surg. Oncol. 65284297
  11. 11. HausteinVSchumacherU2012A dynamic model for tumour growth and metastasis formationJ Clin Bioinforma. 21123
  12. 12. GhotraV. PHeSDe BontHVan Der EntWSpainkH. Pvan de Water B, Snaar-Jagalska BE, Danen EH (2012Automated whole animal bio-imaging assay for human cancer dissemination. PLoS One. 7: e31281.
  13. 13. BaudisM2007Genomic imbalances in 5918 malignant epithelial tumors: an explorative meta-analysis of chromosomal CGH data.BMC Cancer7226241
  14. 14. RørthP2011Whence directionality: guidance mechanisms in solitary and collective cell migrationDev Cell. 20918
  15. 15. RidleyA. JSchwartzM. ABurridgeKFirtelR. AGinsbergM. HBorisyGet al2003Cell migration: integrating signals from front to back.Science. 30217041709
  16. 16. YoshidaKSoldatiT2006Dissection of amoeboid movement into two mechanically distinct modes.J Cell Sci. 119383344
  17. 17. AbrahamV. CKrishnamurthiVTaylorD. LLanniF1999The actin-based nanomachine at the leading edge of migrating cells.Biophys J. 77172132
  18. 18. DunnG. AZichaD1995Dynamics of fibroblast spreading.J Cell Sci. 108123949
  19. 19. NabeshimaKInoueTShimaoYSameshimaT2002Matrix metalloproteinases in tumor invasion: role for cell migrationPathol. Int. 52255264
  20. 20. WolfKMazoILeungHEngelkeKVon AndrianU. HDeryuginaE. Iet al2003Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis.J Cell Biol. 160267277
  21. 21. ArjonenAKaukonenRIvaskaJ2011Filopodia and adhesion in cancer cell motilityCell Adh Migr. 542130
  22. 22. MüllerAHomeyBSotoHGeNCatronDBuchananM. Eet al2001Involvement of chemokine receptors in breast cancer metastasis. Nature. 4105056
  23. 23. LazennecGRichmondA2010Chemokines and chemokine receptors: new insights into cancer-related inflammationTrends Mol. Med. 16133144
  24. 24. YilmazMChristoforiGLehembreF2007Distinct mechanisms of tumor invasion and metastasisTrends Mol Med 13535541
  25. 25. YilmazMChristoforiG2010Mechanisms of motility in metastazising cells. Mol Cancer Res. 8629642
  26. 26. FriedlP2004Prespecification and plasticity: shifting mechanisms of cell migrationCurr Opin Cell Biol. 161423
  27. 27. FriedlPWolfK2010Plasticity of cell migration: a multiscale tuning modelJ Cell Biol. 188119
  28. 28. SahaiE2005Mechanisms of cancer cell invasion.Curr Opin Genet Dev. 158796
  29. 29. ShookDKellerR2003Mechanisms, mechanics and function of epithelial-mesenchymal transitions in early development. Mech Dev 12013511383
  30. 30. CondeelisJSegallJ. E2003Intravital imaging of cell movement in tumours.Nat Rev Cancer. 3921930
  31. 31. BrundageR. AFogartyK. ETuftR. AFayF. S1991Calcium gradients underlying polarization and chemotaxis of eosinophils.Science. 254703706
  32. 32. RidleyAJschwartzM. ABurridgeKFirtelR. AGinsbergM. HBorisyGet al2003Cell migration: integrating signals from front to back.Science. 30217041709
  33. 33. PrevarskayaNSkrymaRShubaY2011Calcium in tumour metastasis: new roles for known actors.Nat Rev Cancer. 1160918
  34. 34. WolfKWuY. ILiuYGeigerJTamEOverallCet al2007Multi-step pericellular proteolysis controls the transition from individual to collective cancer cell invasion.Nat Cell Biol. 9893904
  35. 35. ThieryJ. PAcloqueHHuangR. YNietoM. A2009Epithelial-mesenchymal transitions in development and diseaseCell139871890
  36. 36. RørthP2007Collective guidance of collective cell migrationTrends Cell Biol. 17575579
  37. 37. OlsonM. F2010Follow the leader: LIM kinases pave the way for collective tumor cell invasionCell Cycle944178
  38. 38. IlinaOFriedlP2009Mechanisms of collective cell migration at a glance.J Cell Sci. 12232033208
  39. 39. NabeshimaKInoueTShimaoYOkadaYItohYSeikiMKoonoM2000Front-cell-specific expression of membrane-type 1 matrix metalloproteinase and gelatinase A during cohort migration of colon carcinoma cells induced by hepatocyte growth factor/scatter factor.Cancer Res. 6033649
  40. 40. FriedlPWolfK2008Tube travel: the role of proteases in individual and collective cancer cell invasion.Cancer Res. 6872479
  41. 41. González-alvaPTanakaAOkuYMiyazakiYOkamotoEFujinamiMet al2010Enhanced expression of podoplanin in ameloblastomas. J Oral Pathol Med. 391039
  42. 42. GaggioliCHooperSHidalgo-carcedoCGrosseRMarshallJ. FHarringtonKet al2007Fibroblastled collective invasion of carcinoma cells with differing roles for RhoGTPases in leading and following cells. Nat Cell Biol. 913921400
  43. 43. WickiALehembreFWickNHantuschBKerjaschkiDChristoforiG2006Tumor invasion in the absence of epithelial-mesenchymal transition: podoplanin mediated remodeling of the actin cytoskeletonCancer Cell9261272
  44. 44. Harris TJCTepass U (2010Adherens junctions: from molecules to morphogenesisNat Rev Mol Cell Biol. 11502514
  45. 45. TakeichiM1991Cadherin cell adhesion receptors as a morphogenetic regulator.Science. 25114511455
  46. 46. ShenLWeberC. RRaleighD. RYuDTurnerJ. R2011Tight junction pore and leak pathways: a dynamic duo.Annu Rev Physiol. 73283309
  47. 47. HartsockANelsonW. J2008Adherens and tight junctions: structure, function and connections to the actin cytoskeleton.Biochim Biophys Acta. 1778660669
  48. 48. BaumBGeorgiouM2011Dynamics of adherens junctions in epithelial establishment, maintenance, and remodeling. J Cell Biol. 192907917
  49. 49. TurnerJ. R2009Intestinal mucosal barrier function in health and diseaseNat Rev Immunol. 9799809
  50. 50. Lillie, FR (1908) The Development of the Chick. Henry Holt and Co, New York.
  51. 51. Vincent-salomonAThieryJ. P2003Host microenvironment in breast cancer development: epithelial-mesenchymal transition in breast cancer development.Breast Cancer Res. 5101106
  52. 52. KalluriRWeinbergR. A2009The basics of epithelial-mesenchymal transitionJ Clin Invest. 11914208
  53. 53. ThieryJ. P2002Epithelial-mesenchymal transitions in tumour progression.Nat Rev Cancer. 244254
  54. 54. BatlleESanchoEFranciCDominguezDMonfarMBaulidaJet al2000The transcription factor snail is a repressor of E-cadherin gene expression in epithelial tumour cells.Nat Cell Biol. 28489
  55. 55. YangJWeinbergR. A2008Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasisDev Cell. 1481829
  56. 56. KangYMassagueJ2004Epithelial-mesenchymal transitions: twist in development and metastasis. Cell1182779
  57. 57. Adams CL & Nelson WJ (1998Cytomechanics of cadherinmediated cell-cell adhesionCurr Opin Cell Biol. 10572577
  58. 58. MaedaMJohnsonK. RWheelockM. J2005Cadherin switching: essential for behavioral but not morphological changes during an epithelium-tomesenchyme transition. J Cell Sci. 118873887
  59. 59. NakajimaSDoiRToyodaETsujiSWadaMKoizumiMet al2004N-cadherin expression and epithelial-mesenchymal transition in pancreatic carcinoma. Clin Cancer Res 1041254133
  60. 60. CamandEPeglionFOsmaniNSansonMEtienne-mannevilleS2012N-cadherin expression level modulates integrin-mediated polarity and strongly impacts on the speed and directionality of glial cell migration.J Cell Sci. Jan 24.
  61. 61. GuptaS. KOommenSAubryM. CWilliamsB. PVlahakisN. E2012Integrinapromotes malignant tumor growth and metastasis by potentiating epithelial-mesenchymal transition. Oncogene. Feb 27. doi:onc.2012.41.
  62. 62. ThisseBEl MessalMPerrin-schmittF1987The twist gene: isolation of a Drosophila zygotic gene necessary for the establishment of dorsoventral pattern.Nucleic Acids Res. 1534393453
  63. 63. YangJManiS. ADonaherJ. LRamaswamySItzyksonR. AComeCet al2004Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 117927939
  64. 64. YangW. HLanH. YHuangC. HTaiS. KTzengC. HKaoS. Yet al2012RAC1 activation mediates Twist1induced cancer cell migration.Nat Cell Biol. Mar 11. doi:ncb2455.
  65. 65. PinoM. SBalsamoMDi Modugno F, Mottolese M, Alessio M, Melucci E, et al. (2008Human Mena+11a isoform serves as a marker of epithelial phenotype and sensitivity to epidermal growth factor receptor inhibition in human pancreatic cancer cell lines. Clin Cancer Res. 1449434950
  66. 66. YeoG. WCoufalN. GLiangT. YPengG. EFuX. DGageF. H2009An RNA code for the FOX2 splicing regulator revealed by mapping RNA-protein interactions in stem cellsNat Struct Mol Biol. 16130137
  67. 67. WarzechaC. CJiangPAmirikianKDittmarK. ALuHShenSet al2010An ESRPregulated splicing programme is abrogated during the epithelial-mesenchymal transition. Embo J. 2932863300
  68. 68. ShapiroI. MChengA. WFlytzanisN. CBalsamoMCondeelisJ. SOktayM. Het al2011An EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype.PLoS Genet. 7: e1002218 EOF
  69. 69. TiwariNGheldofATatariMChristoforiG2012EMT as the ultimate survival mechanism of cancer cellsSemin Cancer Biol. Mar 8.
  70. 70. ChaudhuryAHusseyG. SHoweP. H2011UTR-mediated post-transcriptional regulation of cancer metastasis: beginning at the end. RNA Biol. 85959
  71. 71. HusseyG. SChaudhuryADawsonA. ELindnerD. JKnudsenC. RWilceM. Cet al2011Identification of an mRNP complex regulating tumorigenesis at the translational elongation stepMol Cell. 4141931
  72. 72. SunYWangB. ELeongK. GYuePLiLJhunjhunwalaSet al2011Androgen Deprivation Causes Epithelial-Mesenchymal Transition in the Prostate: Implications for Androgen-deprivation Therapy.Cancer Res. 2011 Nov 22.
  73. 73. ParkS. MGaurA. BLengyelEPeterM. E2008The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2Genes Dev. 22894907
  74. 74. GregoryP. ABertA. GPatersonE. LBarryS. CTsykinAFarshidGet al2008The miR- 200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIPNat Cell Biol. 10593601
  75. 75. RuPSteeleRNewhallPPhillipsN. JTothKRayR. B2012MicroRNA-29b Suppresses Prostate Cancer Metastasis by Regulating Epithelial-Mesenchymal Transition Signaling. Mol Cancer Ther. Mar 8.
  76. 76. ForoniCBrogginiMGeneraliDDamiaG2011Epithelial-mesenchymal transition and breast cancer: Role, molecular mechanismsand clinical impact.Cancer Treat Rev. Nov 25.
  77. 77. WellsAYatesCShepardC. R2008E-cadherin as an indicator of mesenchymal to epithelial reverting transitions during the metastatic seeding of disseminated carcinomasClin Exp Metastasis. 256218
  78. 78. LeontovichA. AZhangSQuatraroCIankovIVerouxP. FGambinoM. WDet al. (2012Raf-1 oncogenic signaling is linked to activation of mesenchymal to epithelial transition pathway in metastatic breast cancer cellsInt J Oncol. Mar 19.
  79. 79. PinhoS. SOliveiraPCabralJCarvalhoSHuntsmanDGärtnerFet al2011Loss and Recovery of Mgat3 and GnT-III Mediated E-cadherin N-glycosylation Is a Mechanism Involved in Epithelial-Mesenchymal-Epithelial Transitions.PLoS Onee33191 EOF
  80. 80. HurstD. REdmondsM. DWelchD. R2009bMetastamirthe field of metastasis-regulatory microRNA is spreading. Cancer Res. 6974957498
  81. 81. WangLWangJ2011MicroRNA-mediated breast cancer metastasis: from primary site to distant organs.OncogeneOct 3. doi:onc.2011.444.
  82. 82. KorpalMLeeE. SHuGKangY2008The miR-200 family inhibits epithelial-mesenchymal transition and cancer cell migration by direct targeting of E-cadherin transcriptional repressors ZEB1 and ZEB2J Biol Chem. 2831491014914
  83. 83. LvX. BJiaoYQingYHuHCuiXLinTet al2011miR-124 suppresses multiple steps of breast cancer metastasis by targeting a cohort of pro-metastatic genes in vitro.Chin J Cancer. 3082130
  84. 84. HegerfeldtYTuschMBröckerE. BFriedlP2002Collective cell movement in primary melanoma explants: plasticity of cell-cell interaction, beta1-integrin function, and migration strategies.Cancer Res. 62212530
  85. 85. WolfKMazoILeungHEngelkeKVon AndrianU. HDeryuginaE. Iet al2003Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis.J. Cell Biol. 160267277
  86. 86. ChambersA. FGroomA. CMacDonald IC (2002Dissemination and growth of cancer cells in metastatic sites.Nature Rev. Cancer. 2563572
  87. 87. DvorakH. FWeaverV. MTlstyT. DBergersG2011Tumor microenvironment and progression.J Surg Oncol.103: 468e74.
  88. 88. RoweR. GWeissS. J2008Breaching the basement membrane: who, when and how?Trends Cell Biol. 18560574
  89. 89. HotaryKLiX. YAllenEStevensS. LWeissS. J2006A cancer cell metalloprotease triad regulates the basement membrane transmigration programGenes Dev. 2026732686
  90. 90. TimplRBrownJ. C1996Supramolecular assembly of basement membranes.Bioessays. 18123132
  91. 91. WangSVoisinM. BLarbiK. YDangerfieldJScheiermannCTranMet al2006Venular basement membranes contain specific matrix protein low expression regions that act as exit points for emigrating neutrophilsJ Exp Med. 20315191532
  92. 92. PedersenJ. ASwartzM. A2005Mechanobiology in the third dimensionAnn Biomed Eng. 33: 1469e90.
  93. 93. EnglerA. JCarag-kriegerCJohnsonC. PRaabMTangH. YSpeicherD. Wet al2008Embryonic cardiomyocytes beat best on a matrix with heart-like elasticity: scar-like rigidity inhibits beatingJ Cell Sci. 12137943802
  94. 94. FriedlandJ. CLeeM. HBoettigerD2009Mechanically activated integrin switch controls alpha5beta1 function.Science. 323642644
  95. 95. SchwartzM. A2001Integrin signaling revisitedTrends Cell Biol. 11466470
  96. 96. IndraIBeningoK. A2011An in vitro correlation of metastatic capacity, substrate rigidity, and ECM composition.J Cell Biochem. 11231518
  97. 97. LeventalK. RYuHKassLLakinsJ. NEgebladMErlerJ. TFongS. FCsiszarKGiacciaAWeningerWet alMatrix crosslinking forces tumor progression by enhancing integrin signalingCell2009891 EOF906 EOF
  98. 98. SKumarWeaverV2009Mechanics, malignancy, and metastasis: the force journey of a tumor cell, Cancer Metastasis Rev. 2811327
  99. 99. BrentnallT. ALaiL. AColemanJBronnerM. PPanSChenR2012Arousal of cancer-associated stroma: overexpression of palladin activates fibroblasts to promote tumor invasion.PLoS Onee30219 EOF
  100. 100. MalanchiISantamaria-martínezASusantoEPengHLehrH. ADelaloyeJ. FHuelskenJ2011Interactions between cancer stem cells and their niche govern metastatic colonizationNature. 481859
  101. 101. MøllerH. DRalfkjærUCremersNFrankelMPedersenR. TKlingelhöferJet al2011Role of fibulin-5 in metastatic organ colonization. Mol Cancer Res. 955363
  102. 102. CarasITucureanuCLerescuLPiticaRMelinceanuLNeaguSet al2011Influence of tumor cell culture supernatants on macrophage functional polarization: in vitro models of macrophage-tumor environment interaction.Tumori9764754
  103. 103. SicaAPortaCRiboldiELocatiM2010Convergent pathways of macrophage polarization: The role of B cellsEur J Immunol. 4021313
  104. 104. SicaALarghiPMancinoARubinoLPortaCTotaroM. Get al2008Macrophage polarization in tumour progression. Sem Cancer Biol. 18349355
  105. 105. RomerJNielsenB. SPlougM2004The urokinase receptor as a potential target in cancer therapy.Curr Pharm Des. 1023592376
  106. 106. LazennecGRichmondA2010Chemokines and chemokine receptors: new insights into cancer-related inflammationTrends Mol Med. 1613344
  107. 107. WyckoffJWangWLinE. YWangYPixleyFStanleyE. Ret al2004A paracrine loop between tumor cells and macrophages is required for tumor cell migration in mammary tumors.Cancer Res. 6470227029
  108. 108. CampbellM. JTonlaarN. YGarwoodE. RHuoDMooreD. HKhramtsovA. Iet al2010Proliferating macrophages associated with high grade, hormone receptor negative breast cancer and poor clinical outcomeBreast Cancer Res Treat. 12870311
  109. 109. HembruffS. LJokarIYangLChengN2010Loss of transforming growth factor-beta signaling in mammary fibroblasts enhances CCL2 secretion to promote mammary tumor progression through macrophage-dependent and-independent mechanisms. Neoplasia. 1242533
  110. 110. TanWZhangWStrasnerAGrivennikovSChengJ. QHoffmanR. Met al2011Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling.Nature47054853
  111. 111. DenardoD. GBarretoJ. BAndreuPVasquezLTawfikDKolhatkarNet al2009CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell1691102
  112. 112. LiNKarinM1999IsN. F-k. a. p. aB the sensor of oxidative stress? FASEB J. 1311371143
  113. 113. PanteleakouZLembessisPSourlaAPissimissisNPolyzosADeliveliotisCet al2009Detection of circulating tumor cells in prostate cancer patients: methodological pitfalls and clinical relevanceMol Med. 15101114
  114. 114. SakamotoSMccannR. ODhirRKyprianouN2010Talin1 promotes tumor invasion and metastasis via focal adhesion signaling and anoikis resistanceCancer Res. 70188595
  115. 115. HalderGJohnsonR. L2011Hippo signaling. Growth control and beyondDevelopment. 138922
  116. 116. ZhaoBLiLWangLWangC. YYuJGuanK. L2012Cell detachment activates the Hippo pathway via cytoskeleton reorganization to induce anoikisGenes Dev. 265468
  117. 117. LamarJ. MSternPLiuHSchindlerJ. WJiangZ. GHynesR. O2012The Hippo pathway target, YAP, promotes metastasis through its TEAD-interaction domainProc Natl Acad Sci U S A. 109: E2441E2450.
  118. 118. KroemerGEl-DeiryW. SGolsteinPPeterM. EVauxDVandenabeelePet al2005Classification of cell death: recommendations of the Nomenclature Committee on Cell DeathCell Death Differ. 1214631467
  119. 119. GalluziLMaiuriM. CVitaleIZischkaHCastedoMZitvogelLet al2007Cell death modalities: classification and pathophysiological implications. Cell Death Differ. 1412371266
  120. 120. WongR2011Apoptosis in cancer: from pathogenesis to treatmentJ Exp Clin Cancer Res. 3087100
  121. 121. KumarRVadlamudiR. KAdamL2000Apoptosis in mammary gland and cancer.Endocr Relat Cancer. 7257269
  122. 122. GiannoniEBuricchiFGrimaldiGParriMCialdaiFTaddeiM. Let al2008Redox regulation of anoikis: reactive oxygen species as essential mediators of cell survivalCell Death Differ. 15867878
  123. 123. Eisenberg-lernerABialikSSimonH. UKimchiA2009Life and death partners: apoptosis, autophagy and the cross-talk between themCell Death Differ. 16966975
  124. 124. SimpsonC. DAnyiweKSchimmerD2008Anoikis resistance and tumor metastasisCancer Lett. 272177185
  125. 125. GuadamillasM. CCerezoADel Pozo MA (2011Overcoming anoikis--pathways to anchorage-independent growth in cancer.J Cell Sci. 124318997
  126. 126. PuthalakathHHuangD. COReillyL. AKingS. MStrasserA (1999The proapoptotic activity of the bcl-2 family member bim is regulated by interaction with the dynein motor complexMol Cell. 328796
  127. 127. LoftusJ. CLiddingtonR. C1997Cell adhesion in vascular biology. New insights into integrin-ligand interaction.J Clin Invest. 9923026
  128. 128. GilmoreA. P2005Anoikis. Cell Death Differ. 12(Suppl. 2): 1473-1477.
  129. 129. HynesR. O2002ntegrins: bidirectional, allosteric signaling machines. Cell. 11067387
  130. 130. AlahariS. KReddigP. JJulianoR. L2002Biological aspects of signal transduction by cell adhesion receptors.Int Rev Cytol. 220145184
  131. 131. HanksS. KRyzhovaLShinN. YBrábekJ2003Focal adhesion kinase signaling activities and their implications in the control of cell survival and motility.Front Biosci. 8: d982996
  132. 132. CalalbM. BPolteT. RHanksS. K1995Tyrosine phosphorylation of focal adhesion kinase at sites in the catalytic domain regulates kinase activity: a role for Src family kinases.Mol Cell Biol. 1595463
  133. 133. IlicDAlmeidaE. ASchlaepferD. DDazinPAizawaSDamskyC. H1998Extracellular matrix survival signals transduced by focal adhesion kinase suppress 53apoptosis.J Cell Biol. 143: 547-560.
  134. 134. HorbinskiCMojeskyCKyprianouN2010Live free or die: tales of homeless (cells) in cancer.Am J Pathol. 177104452
  135. 135. CardoneM. HRoyNStennickeH. RSalvesenG. SFrankeT. FStambridgeEet al1998Regulation of cell death protease caspase-9 by phosphorylation. Science. 28213181321
  136. 136. DattaS. RKatsovAHuLFesikS. WYaffeM. BGreenbergM. E200014-3-3 proteins and survival kinases cooperate to inactivate BAD by BH3 domain phosphorylationMol Cell. 64151
  137. 137. RomashkovaJ. AMakarovS. S1999N. F-?BIsa target of AKT in antiapoptotic PDGF signalling. Nature. 4018690
  138. 138. KopsG. JBurgeringB. M1999Forkhead transcription factors: new insights into protein kinase B (c-akt) signaling.J Mol Med. 77656665
  139. 139. DuxburyM. SItoHZinnerM. JAshleyS. WWhangE. E2004Focal adhesion kinase gene silencing promotes anoikis and suppresses metastasis of human pancreatic adenocarcinoma cellsSurgery13555562
  140. 140. GoundiamONagelM. DVayssadeM2012Akt and RhoA inhibition promotes anoikis of aggregated B16F10 melanoma cellsCell Biol Int. 363119
  141. 141. CarterW. GWaynerE. ABouchardT. SKaurP1990The role of integrins a2ß1 and a3ß1 in cell-cell and cell-substrate adhesion of human epidermal cells. J Cell Biol. 11013871404
  142. 142. BreussJ. MGalloJDelisserH. MKlimanskayaI. VFolkessonH. GPittetJ. Fet al1995Expression of the ß6 integrin subunit in development, neoplasia and tissue repair suggests a role in epithelial remodeling. J Cell Sci. 10822412251
  143. 143. HaapasalmiKZhangKTonnesenMOlerudJSheppardDSaloTet al1996Keratinocytes in human wounds express avß6 integrin. J Invest Dermatol. 1064248
  144. 144. RegeziJ. ARamosD. MPytelaRDekkerN. PJordanN. C2002Tenascin and ß6 integrin are overexpressed in floor of mouth in situ carcinomas and invasive squamous cell carcinomas. Oral Oncol. 38332336
  145. 145. MontgomeryA. MReisfeldR. AChereshD. A1994Integrin avß3 rescues melanoma cells from apoptosis in three-dimensional dermal collagen. Proc Natl Acad Sci USA. 9188568860
  146. 146. GoldsteinN. BJohannesW. UGadeliyaA. VGreenM. RFujitaMNorrisD. Aet al2009Active N-Ras and B-Raf inhibit anoikis by down-regulating Bim expression in melanocytic cells. J Invest Dermatol. 1294327
  147. 147. Albelda SM, MetteSA, Elder DE, Stewart R, Damjanovich L, Herlyn M, et al (1990) Integrin distribution in malignant melanoma: association of the beta 3 subunit with tumor progression. Cancer Res. 50: 6757-6764.
  148. 148. KozlovaN. IMorozevichG. EChubukinaA. NBermanA. E2001Integrin alphavbeta3 promotes anchorage-dependent apoptosis in human intestinal carcinoma cells.Oncogene2047104717
  149. 149. ZhuPTanM. JHuangR. LTanC. KChongH. CPalMet al2011Angiopoietin-like 4 protein elevates the prosurvival intracellular O2(-):H2O2 ratio and confers anoikis resistance to tumors. Cancer Cell. 19401415
  150. 150. ShenWChenDFuHLiuSSunKSunX2011S100A4 protects gastric cancer cells from anoikis through regulation of av and a5 integrin. Cancer Sci. 10210148
  151. 151. MarchánSPérez-torrasSVidalAAdanJMitjansFCarbóNet al2011Dual effects of ß3 integrin subunit expression on human pancreatic cancer models. Cell Oncol (Dordr). 34393405
  152. 152. TsengYLeeJ. SKoleT. PJiangIWirtzD2004Micro-organization and visco-elasticity of the interphase nucleus revealed by particle nanotracking.J Cell Sci. 117215967
  153. 153. FidlerI. JYanoSZhangR. DFujimakiTBucanaC. D2002The seed and soil hypothesis: vascularisation and brain metastasesLancet Oncol. 3537
  154. 154. WeissLOrrF. WHonnK. V1988Interactions of cancer cells with the microvasculature during metastasis.FASEB J. 21221
  155. 155. MengSTripathyDFrenkelE. PSheteSNaftalisE. ZHuthJ. Fet al2004Circulating tumor cells in patients with breast cancer dormancy.Clin Cancer Res. 10815262
  156. 156. PngK. JHalbergNYoshidaMTavazoieS. F2011A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells.Nature4811904
  157. 157. BrownD. MRuoslahtiE2004Metadherin, a cell surface protein in breast tumors that mediates lungmetastasis.Cancer Cell536574
  158. 158. EylesJPuauxA. LWangXTohBPrakashCHongMet al2010Tumor cells disseminate early, but immunosurveillance limits metastatic outgrowth, in a mouse model of melanomaJ Clin Invest. 12020302039
  159. 159. LeongS. PNakakuraE. KPollockRChotiM. AMortonD. LHennerW. Det al2011Unique patterns of metastases in common and rare types of malignancy.J Surg Oncol. 103607614
  160. 160. KienastYVon BaumgartenLFuhrmannMKlinkertW. EGoldbrunnerRHermsJet al2010Real-time imaging reveals the single steps of brain metastasis formation. Nature Med. 16116122
  161. 161. PalumboJ. STalmageK. EMassariJ. VLa Jeunesse CM, Flick MJ, Kombrinck KW, et al. (2005Platelets and fibrin(ogen) increase metastatic potential by impeding natural killer cellmediated elimination of tumor cells. Blood. 105178185
  162. 162. NashGTurnerLScullyMKakkarA2002Platelets and cancer. Lancet Oncol. 3425430
  163. 163. NicolsonG. L1988Cancer metastasis: TC and host organ properties important in metastasis to specific secondary sites. Biochem Biophys Acta. 948175224
  164. 164. PagetS1889The distribution of secondary growths in cancer of the breast.Lancet. 1571573
  165. 165. PsailaBKaplanR. NPortE. RLydenD2007Priming the’soil’ for breast cancer metastasis: the pre-metastatic niche.Breast Dis. 266574
  166. 166. PerelmuterV. MManskikhV. N2012Preniche as Missing Link of the Metastatic Niche Concept Explaining Organ_Preferential Metastasis of Malignant Tumors and the Type of Metastatic DiseaseBiochemistry7711118
  167. 167. AliSLazennecG2007Chemokines novel targets for breast cancer metastasis. Cancer Metastasis Rev. 26401420
  168. 168. Baruch-benA2008Organ selectivity in metastasis: regulation by chemokines and their receptorsClin Exp Metastasis. 25345356
  169. 169. WendelCHemping-bovenkerkAKrasnyanskaJMeesS. TKochetkovaMStoeppelerSet al2012CXCR4/CXCL12 participate in extravasation of metastasizing breast cancer cells within the liver in a rat model.PLoS Onee30046 EOF
  170. 170. InoueSOsmondD. G2001Basement membrane of mouse bone marrow sinusoids shows distinctive structure and proteoglycan composition: a high resolution ultrastructural study.Anat Rec. 264294304
  171. 171. WeilR. JPalmieriD. CBronderJ. LStarkA. MSteegP. S2005Breast cancer metastasis to the central nervous systemAm J Pathol. 167913920
  172. 172. LeeY. T1985Patterns of metastasis and natural courses of breast carcinoma.Cancer Metastasis Rev. 4153172
  173. 173. JohanssonJ. EAndrénOAnderssonS. ODickmanP. WHolmbergLMagnusonAet al2004Natural history of early, localized prostate cancer. JAMA. 29127132719
  174. 174. TriozziP. LEngCSinghA. D2008Targeted therapy for uveal melanomaCancer Treat. Rev. 34247258
  175. 175. Nieto J, Grossbard ML, Kozuch P (2008) Metastatic pancreatic cancer 2008: is the glass less empty? Oncologist. 13 562576 .
  176. 176. FeldRRubinsteinL. VWeisenbergerT. H1984Sites of recurrence in resected stage I non-small-cell lung cancer: a guide for future studies.J. Clin. Oncol. 213521358
  177. 177. Aguirre-ghisoJ. A2007Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 7834846
  178. 178. WeissL1992Comments on hematogenous metastatic patterns in humans as revealed by autopsy.Clin Exp Metastasis 10191199
  179. 179. GassmannPHaierJ2008The tumor cell-host organ interface in the early onset of metastatic organ colonization. Clin. Exp. Metastasis. 25171181
  180. 180. Barnholtz-sloanJ. SSloanA. EDavisF. GVigneauF. DLaiPSawayaR. E2004Incidence proportions of brain metastases in patients diagnosed in the Metropolitan Detroit Cancer Surveillance System. J Clin Oncol.2228652872
  181. 181. DelattreJ. YKrolGThalerH. T1988Distribution of brain metastases.Arch Neurol. 45741744
  182. 182. EichlerA. FKuterIRyanPSchapiraLYoungerJHensonJ. Wet al2008Survival in patients with brain metastases from breast cancer: the importance of HER-2 statusCancer11223592367
  183. 183. KangYSiegelP. MShuWDrobnjakMKakonenS. MCordon-cardoCet alA multigenic program mediating breast cancer metastasis to boneCancer Cell353749
  184. 184. RoodmanG. D2004Mechanisms of bone metastasis.N Engl J Med. 350165564
  185. 185. IbrahimTFlaminiEMercataliLSacannaESerraPAmadoriD2010Pathogenesis of osteoblastic bone metastases from prostate cancer. Cancer. 116140618
  186. 186. BosP. DZhangX. HNadalCShuWGomisR. RNguyenD. Xet al2009Genes that mediate breast cancer metastasis to the brain. Nature. 45910059
  187. 187. HengelKSidhuGChoiJWeedonJNwokediEAxiotisC. Aet al2012Attributes of brain metastases from breast and lung cancerInt J Clin Oncol. Mar 2.
  188. 188. LalorP. FEdwardsSMcnabGSalmiMJalkanenSAdamsD. H2002Vascular adhesion protein-1 mediates adhesion and transmigration of lymphocytes on human hepatic endothelial cells.J Immunol. 16998392

Written By

M.E. Hernández-Caballero

Submitted: 29 November 2011 Published: 23 January 2013