Next Article in Journal
Association between Physiological and Subjective Aspects of Pain and Disability in Post-Stroke Patients with Shoulder Pain: A Cross-Sectional Study
Next Article in Special Issue
Deciphering Hydrodynamic and Drug-Resistant Behaviors of Metastatic EMT Breast Cancer Cells Moving in a Constricted Microcapillary
Previous Article in Journal
Clusters of Glycemic Response to Oral Glucose Tolerance Tests Explain Multivariate Metabolic and Anthropometric Outcomes of Bariatric Surgery in Obese Patients
Previous Article in Special Issue
ATP Synthase Subunit Epsilon Overexpression Promotes Metastasis by Modulating AMPK Signaling to Induce Epithelial-to-Mesenchymal Transition and Is a Poor Prognostic Marker in Colorectal Cancer Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Markers of Cancer Cell Invasion: Are They Good Enough?

by
Tatiana S. Gerashchenko
1,*,
Nikita M. Novikov
1,2,
Nadezhda V. Krakhmal
3,
Sofia Y. Zolotaryova
2,
Marina V. Zavyalova
3,4,
Nadezhda V. Cherdyntseva
1,5,
Evgeny V. Denisov
1,6 and
Vladimir M. Perelmuter
4
1
Laboratory of Molecular Oncology and Immunology, Cancer Research Institute, Tomsk National Research Medical Center, 634009 Tomsk, Russia
2
Department of Cytology and Genetics, Tomsk State University, 634050 Tomsk, Russia
3
Department of Pathological Anatomy, Siberian State Medical University, 634050 Tomsk, Russia
4
Department of General and Molecular Pathology, Cancer Research Institute, Tomsk National Research Medical Center, 634009 Tomsk, Russia
5
Laboratory for Translational Cellular and Molecular Biomedicine, Tomsk State University, 634050 Tomsk, Russia
6
Department of Organic Chemistry, Tomsk State University, 634050 Tomsk, Russia
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2019, 8(8), 1092; https://doi.org/10.3390/jcm8081092
Submission received: 28 June 2019 / Revised: 20 July 2019 / Accepted: 22 July 2019 / Published: 24 July 2019

Abstract

:
Invasion, or directed migration of tumor cells into adjacent tissues, is one of the hallmarks of cancer and the first step towards metastasis. Penetrating to adjacent tissues, tumor cells form the so-called invasive front/edge. The cellular plasticity afforded by different kinds of phenotypic transitions (epithelial–mesenchymal, collective–amoeboid, mesenchymal–amoeboid, and vice versa) significantly contributes to the diversity of cancer cell invasion patterns and mechanisms. Nevertheless, despite the advances in the understanding of invasion, it is problematic to identify tumor cells with the motile phenotype in cancer tissue specimens due to the absence of reliable and acceptable molecular markers. In this review, we summarize the current information about molecules such as extracellular matrix components, factors of epithelial–mesenchymal transition, proteases, cell adhesion, and actin cytoskeleton proteins involved in cell migration and invasion that could be used as invasive markers and discuss their advantages and limitations. Based on the reviewed data, we conclude that future studies focused on the identification of specific invasive markers should use new models one of which may be the intratumor morphological heterogeneity in breast cancer reflecting different patterns of cancer cell invasion.

1. Introduction

Metastasis is a key feature of cancer and a “final chord” of the tumor progression [1]. The ability for metastasis enables tumor cells to leave the primary site and disseminate throughout the body, causing severe organ failure and leading to death. Understanding the mechanisms underlying metastasis is extremely important for the development of highly effective cancer therapies [2].
Metastasis is a complex process of stepwise events collectively termed the metastatic cascade and consisting of local invasion of tumor cells, intravasation to blood vessels, survival in the circulation, arrest at distant organs, extravasation into the parenchyma of distant tissues, micrometastasis formation, and metastatic colonization (macrometastasis) [1,2]. Invasion is the first step in the metastasis of tumor cells. From the morphological point of view, the invasion is a process during which malignant cells detach from the tumor mass, acquire the ability to actively move, and invade surrounding tissues through the adjacent basement membrane [3]. The interface of tumor and host tissue, in other words, the deepest rim of cancerous tissue grown in adjacent non-cancerous tissues, is called an “invasive front (edge)” [4]. Tumor cells constituting the invasive front are phenotypically different from cells in other tumor parts. Invasive front cells are believed to have a locomotor phenotype and demonstrate a variety of types and mechanisms of movement [5,6]. Tumor cells can move collectively or individually. The type of invasion depends on the molecular changes in tumor cells and the tumor microenvironment features [7,8,9,10]. The distinctive features of collective cell invasion include physical and functional relationships among tumor cells due to adhesion molecules as well as the presence of leader cells that are characterized by the mesenchymal phenotype and the ability to form lamellipodia, pull follower cells, and destroy the extracellular matrix (ECM) through production of proteases [11,12,13]. Interestingly, according some reports, invasive leaders do not express molecular features of epithelial–mesenchymal transition (EMT) [14], but exhibit a basal epithelial gene program, that is enriched in cytokeratin-14 and the transcription factor p63 [15,16].
Individual invasion can occur through mesenchymal and amoeboid cell migration mechanisms [17]. Sometimes, an intermediate amoeboid/mesenchymal (filopodial) cell migration mode is distinguished [18]. In mesenchymal movement, tumor cells exhibit a pronounced fibroblast-like phenotype, high expression of integrins, synthesis of proteolytic enzymes, and activity of small GTPases Rac1 and Cdc42 that are necessary to form lamellipodia and actomyosin contractions [7,12]. In amoeboid movement, cells are not capable of proteolysis and adhesion of the ECM but demonstrate the enhanced activity of the actomyosin machinery and the formation of cell membrane protrusions (blebs), which allow cells to squeeze through tight spaces in the surrounding matrix. Amoeboid movement directly depends on Rho/ROCK cell signaling and activity of type II myosin [13,17,19]. Tumor cells can transit from one cell migration phenotype to another via mesenchymal–amoeboid (MAT) and amoeboid–mesenchymal transition. The key role in these transitions is played by the balance of GTPases Rho and Rac, changes in expression of focal adhesion molecules and proteases, and ECM stiffness [13]. Importantly, the Rho/Rac feedback loop, particularly balanced relative high RhoA and Rac1, is also responsible for the hybrid amoeboid/mesenchymal phenotype in migrating cells [20].
EMT plays a key role in tumor dissemination. During EMT, tumor cells lose the epithelial phenotype and acquire the mesenchymal features and resistance to antitumor treatment; EMT also promotes immortalization and is involved in the prevention of apoptosis [21,22]. EMT is induced not only by molecular changes in tumor cells but also by cytokines and growth factors secreted by immune and stromal cells of the tumor microenvironment [23,24,25,26]. EMT may be incomplete (partial) when tumor cells still retain epithelial features but already acquire mesenchymal traits. During partial EMT, cells are described as a hybrid, with an intermediate epithelial/mesenchymal phenotype [27]. Partial EMT has been reported for both single tumor cells and tumor buds (groups of up to five cells) that are a variant of collective invasion [28]. The phenomenon “tumor budding” is regarded as a specific “signal” indicating the onset of cancer invasion and metastasis. The presence of tumor buds in the invasive front was found to be associated with increased metastasis and poor prognosis in various cancers [28,29,30,31,32,33,34].
Tumor cells can acquire the ability for migration not only through EMT but also through the so-called collective–amoeboid transition (CAT) when cells detach from the tumor mass and acquire an amoeboid phenotype rather than a mesenchymal phenotype. CAT is known to be regulated by the core regulatory circuits underlying EMT (miR-200/miR-34) and MAT (Rac1/RhoA) [35] and can be promoted by hypoxia-inducible factor 1 (HIF-1), which is accompanied by a decrease in E-cadherin expression [36]. However, CAT still remains a poorly understood phenomenon.
Active migration of tumor cells is not the only mechanism for invasive tumor growth. There is the so-called passive invasion when cells penetrate adjacent tissues under pressure from other tumor cells during proliferation (expansive growth) or due to an increase in the ECM density caused by the production of fibronectin and collagen by cancer-associated fibroblasts [37,38]. The fact that many circulating tumor cells are apoptotic [39,40], may be considered as indirect evidence of passive invasion, whereas active invasion is associated with viable cells [37].
Despite the fact that the mechanisms and types of cell migration and invasion have been described and studied quite well, there are currently no highly efficient and validated molecular markers for identification of migrating/invading tumor cells in tumors and, therefore, for assessment of their invasive potential. These markers could be used to identify patients at the high risk of distant metastasis and to prescribe therapy aimed at interrupting the metastatic process. In addition, these markers might represent targets for future therapeutics that block invasion and metastasis.
In this review, we systematized information about molecules that might be potential markers of tumor invasion and discussed the advantages and limitations of their use in clinical practice.

2. Potential Markers of Cancer Cell Invasion

The literature reports numerous studies describing various molecules that may act as markers of tumor cell invasion. Conventionally, they may be subdivided into several groups: ECM components, EMT, cell–cell and cell–ECM molecules, proteases, and actin cytoskeleton proteins (Table 1).

2.1. ECM Components

The first barrier to tumor cell invasion is the basement membrane that is a 100–300 nm thick ECM structure consisting of laminins, type IV collagen, and other non-cellular components, on which epithelial cells proliferate and differentiate [41,42,43,44]. Impaired integrity of the basement membrane is a histological marker indicating that carcinoma has acquired invasive properties [12,41,43]. A key component of the basement membrane, laminin-5, consists of α3, β3, and γ2 chains and plays a significant role in migration and invasion of tumor cells [43,45,46,47,48]. The interaction between laminin-5 and tumor cell integrins leads to the release of proteases and degradation of the basement membrane and ECM [43,47,49,50,51]. The laminin-5 γ2 chain monomer, which is considered as one of the most characteristic markers of invasion is found in the invasive front of different cancers [51,52]. For example, laminin γ2 expression combined with MMP-7 and EGFR expression in the invasive front is associated with gastric cancer aggressiveness [43]. In gastric cancer, cytoplasmic expression of laminin γ2 in tumor cells is related to lymph node metastasis and advanced stage [53]; in gallbladder cancer, stromal laminin γ2 expression is associated with a poor prognosis [54]. Laminin γ2 is also expressed in the invasive front of breast, pancreatic, colon, lung, and other cancers [46,51,52,55,56].
After penetrating the basement membrane, invading cells enter the ECM. Fibronectin is the major ECM component that plays a key role in the stimulation of cell growth, adhesion, and cell migration. On the one hand, fibronectin forms a physical barrier for migrating cells; on the other hand, its interaction with tumor cell integrins, mainly with α5β1, triggers ECM proteolysis through secreting MMP-2 and MMP-9 [42,112]. Fibronectin was demonstrated to be involved in the regulation of cell invasion and migration in various cancers [113] and expressed at the invasive front of oral and head and neck squamous cell carcinomas [60,61].
The tenascin C protein also belongs to ECM glycoproteins; however, it is mainly active during embryogenesis. In the adult body, tenascin C is found only in some types of connective tissue (tendons, ligaments, etc.). Interestingly, tenascin C is often expressed in the invasive front of breast, lung, liver, and gallbladder cancers, as well as melanoma, and is associated with a poor prognosis particularly decreased recurrence-free and overall survival and a high rate of metastasis [57,62].
Despite the proven association of basement membrane and ECM components with invasiveness, their role as markers of tumor invasion is ambiguous. For example, laminin γ2 expression is not always observed in the invasive front. According to Sentani [43], cytoplasmic laminin γ2 expression in the invasive front of gastric cancer occurs only in 25% of cases, and stromal expression is observed in 8% of cases. According to García-Solano [58], laminin γ2 expression in tumor buds at the invasive front of colorectal adenocarcinoma is found only in 17–57% of cases. In addition to the invasive front, laminin γ2 is also found in the basement membrane and cytoplasm of tumor cells, outside the invasive front [46]. Fibronectin and tenascin C are also expressed not only in the invasive front [57,59].

2.2. EMT Factors

EMT is common to almost all cancers, but the transition is rarely implemented in full [67]. Partial EMT is mainly typical of tumor cell clusters. However, there is evidence that single migrating cells may be in partial EMT. During partial EMT, tumor cells show co-expression of molecules of epithelial (E-cadherin, EpCAM, cytokeratin 7, miR-200, miR-34, etc.) and mesenchymal (N-cadherin, vimentin, ZEB, SNAIL, etc.) phenotypes. Cells in a partial EMT are capable of both adhesion and migration [27,28,67].
Overexpression of EMT markers is often observed in the invasive front of various cancers [63]. Nevertheless, molecules involved or associated with EMT are characterized by a low diagnostic value in assessing the invasive potential of tumors. Snail and Twist transcription factors are unstable molecules and undergo rapid proteasomal degradation [64,65]. In contrast, according to our data, Snail and Twist are totally expressed in breast tumor, without any selectivity in the invasive front [66]. Vimentin, which is considered a marker of the final EMT stage, may not be expressed in invasive carcinomas at all [67]. Furthermore, EMT is not always necessary for invasion and metastasis. In Snail and Twist knockout mice, tumor dissemination and the number of metastases are comparable to those in control mice [114]. Therefore, the presence of EMT cannot always answer the question whether the tumor cell migrates at a given time.
However, it should be understood that EMT is a complex process in which each step is thought to be regulated by a distinct set of transcription factors and molecular circuits overlapping to each other and generating specific phenotypes [115,116]. The picture is complicated by the fact that EMT transcription factors control other cellular events, including apoptosis and stemness [116]. Moreover, induction of an EMT transcription factor is known to be sufficient to induce single-cell dissemination without orchestrating the molecular EMT program and with retaining epithelial identity [16,117]. Thus, further studies are needed to explore molecular mechanisms underlying each EMT module, namely cell motility, and to find markers that could be used to assess the invasive potential of tumor cells. In addition, it is necessary to consider the fact that cells are capable of amoeboid and hybrid amoeboid/mesenchymal movement. Therefore, a perfect method for determining the invasive phenotype in tumor cells is the simultaneous assessment of markers of mesenchymal and amoeboid migration.

2.3. Cell–Cell and Cell–ECM Interaction Molecules

Adhesion molecules, such as integrins and the cadherin-catenin complex, are the key components of tumor invasion. Changes in the activity of cadherins, which are proteins involved in the formation of cell–cell contacts, is a characteristic feature of invasive growth. E-cadherin, which forms adherens junctions in an epithelial cell layer, is repressed by Snail, Slug, and Twist transcription factors during EMT [64]. The loss of E-cadherin and the nuclear localization of β-catenin, involved in signaling to the actin cytoskeleton [118], were observed in tumor cells at the invasive front in various cancers [69]. Nuclear accumulation of β-catenin in tumor cells in the invasive front and in vessels was found to be a powerful predictor of liver metastasis in colorectal cancer [70,71]. However, the loss of E-cadherin expression is probably not an indispensable prerequisite for invasiveness of tumor cells [72] and, therefore, cannot be used as a marker for invasive growth, at least for some cancers. Moreover, in some tumors, a loss of E-cadherin has been shown to be detrimental to invasion and metastasis. For example, the presence of E-cadherin is a specific feature of a highly aggressive form of breast cancer, inflammatory carcinoma, and needed for successful invasion and metastatic colonization of bone by tumor cells [119]. In this regard, analysis of more effective markers is needed to assess the invasive tumor potential, along with markers of amoeboid movement, as mentioned above.
The key event initiating production of metalloproteinases is the interaction of integrins with ECM components. The main ligands for integrins are fibronectin (α5β1, αvβ3, and α4β1 integrins), collagens (α1β1, α2β1, and α11β1), and laminins (α2β1, α3β1, α6β1, and α6β4) [41,64,120,121,122,123]. For example, α3β1 integrin activates MMP-9 synthesis through interaction with laminins and triggers reorganization of the actin cytoskeleton [124]; α6β1 is involved in tumor invasion via activation of the urokinase plasminogen activator (uPA) receptor and MMP-2 [125]. Laminin-5 is the best-characterized ligand for α3β1 integrin. α6β4 integrin is involved in the regulation of tumor cell migration through activation of the Rho-A signaling cascade [121]. Binding of fibronectin to α5β1 integrin activates MMP-1 and stimulates migration through the ILK/Akt and GSK3β/Snail/E-cadherin signaling pathways [121,126]. Fibronectin-mediated migration is also associated with αvβ3 integrin. αvβ3 integrin is involved in activation of MMP-2 [127] and, under stress conditions, can trigger a ligand-independent signaling cascade leading to activation of NF-κB and Slug, acquisition of a stem phenotype, and promotion of migration [126].
Expression of integrins changes during tumor progression and is often elevated in the invasive front of tumors: αvβ3 in melanoma [75], αvβ6 in colon and head and neck cancers [73,76], and α6β4 in non-small cell lung cancer [74]. Furthermore, high expression of integrins in tumor cells may promote metastasis. For example, α2β1 enhances metastasis of rhabdomyosarcoma in nude mice after intravenous or subcutaneous injection [128], whereas α3β1 promotes lung metastasis through binding to laminin-5 in an exposed basement membrane in the pulmonary vasculature [50].
Signaling pathways activated by different integrins may lead to the same biological effects, while an individual contribution of each of the integrins is different. In neuroblastoma, tumor cell migration can be activated either via FAK-mediated α5β1 integrin signaling or via a FAK-independent pathway involving α4β1 integrin. Both signaling pathways lead to the induction of Src family protein kinases [129,130].
The use of integrins as markers of invasive growth is complicated by the fact that the same integrins can participate in both invasion and other biological processes [78]. For example, α6β1 integrin, apart from involvement in tumor invasion, also participates in Ca2+ signaling [131] and platelet adhesion upon damage to the vascular wall [132].
There is evidence that changes in expression of other cell interaction proteins may be a marker of invasive tumor cells. Galectins, membrane glycoproteins, bound to integrins, laminins, and fibronectin, are used by cells to interact with each other and with the ECM [47,133]. Galectin-1 is involved in the regulation of cell adhesion and migration, on the one hand, through stimulation of MMP-2 and MMP-9 and, on the other hand, through activation of a small Rho GTPase Cdc42, which promotes the formation of actin filopodia. Increased expression of galectin-1 is associated with high invasiveness of lung adenocarcinoma and observed in the invasive front of oral squamous cell carcinoma and glioblastoma [47,79,80]. However, galectins have effects not only on tumor cells but also on immune cells promoting inflammation or dampening T cell-mediated immune responses [77]. The L1 cell adhesion molecule (L1CAM), which is involved in β-catenin/TCF signaling, is necessary for cell migration and invasion. Normally, L1CAM is present only in the nervous tissue, but its expression is induced in tumor cells. Increased expression of L1CAM was found in many cancers, including the invasive front of colorectal and pancreatic cancers [81,82]. Nevertheless, L1CAM can have a static function as a cell adhesion molecule and its expression is associated with good cancer prognosis [83,134].

2.4. Serine Proteases and Matrix Metalloproteinases

One of the main systems responsible for ECM proteolysis is the plasminogen activation system that triggers a powerful serine protease, plasmin. The central component of this system is the uPA and its receptor (uPAR), the interaction of which stimulates proteolysis of plasminogen to plasmin [135,136]. uPA is believed to play a significant role in tumor invasion and metastasis [135,136,137]. Experiments in model animals demonstrated that inhibition of uPA and/or the uPA/uPAR interaction slows down metastasis [135]. In contrast, expression of uPAR is associated with tumor invasion and is found in stromal and tumor cells in the invasive front of oral and skin squamous cell carcinomas [84,85].
Metalloproteinases are involved in proteolytic degradation of the basement membrane and ECM. MMP-7 activates MMP-2 and MMP-9 gelatinases exhibiting proteolytic activity against collagen IV, laminins, proteoglycans, and fibronectin [138]. Expression of MMPs is observed during cancer cell invasion [13,41]. MMP-7-positive tumor cells are predominantly found in the invasive front of gastric cancer, while their number is much higher in aggressive and late-stage tumors [90,91]. MMP-7 is also expressed in the invasive front of colon cancer and correlates with tumor stage [56,91,92]. Elevated MMP-2 and MMP-9 levels are observed in the invasive front of melanoma, endometrial cancer, and ovarian cancer [89,93]. High MMP-2 and MMP-9 expression is also observed in the invasive front of head and neck squamous cell carcinoma [88,94]. Assessment of MMP-2 and MMP-9 expression in the invasive tumor front may be helpful in the differentiation of verrucous carcinoma and squamous cell carcinoma of the oral cavity [139].
However, increased expression of uPA and MMPs is not a unique feature of invasive tumor cells and may be observed in other physiological processes. The components of the uPA system can be involved in the early stages of tumor formation and can increase cell proliferation, inhibit apoptosis, etc. [86]. MMPs are mediators between tumor cells and the microenvironment [87]. MMP-9 produced by inflammatory cells is involved in the proteolytic activation of anti-inflammatory cytokines TGF-β2 and TGF-β3, and MMP-2 and MMP-14 participate in the activation of TGF-β1 [87,140,141]. MMP-2, MMP-9, and MMP-14 indirectly modulate TGF-β activity by cleaving an ECM component, the latent TGF-β binding protein 1 [87,142]. MMP-7 inhibits apoptosis and reduces the efficacy of chemotherapy by cleaving Fas ligands on the surface of cells exposed to doxorubicin [87,143]. MMP-2 and MMP-9 are also involved in the regulation of angiogenesis and lymphangiogenesis [87]. MMP-9 secreted by inflammatory cells modulates bioavailability of VEGF to the VEGFR2 receptor [87,144]. Experiments in mice demonstrated the role of MMP-9 in triggering the angiogenic switch and in vasculogenesis [87,145,146]. Therefore, the multifunctionality of MMPs reduces their significance as markers of invasive growth.

2.5. Actin Cytoskeleton Proteins

Proteins involved in actin cytoskeleton remodeling play an important role in the mechanisms of tumor cell migration and invasion [147]. The ezrin protein is a connecting link between actin filaments and membrane proteins involved in cell–cell adhesion and migration [148]. Ezrin was demonstrated to be localized together with the podoplanin in filopodia, stimulating cellular invasion [149], and expressed in the invasive front of lung cancer [95]. Many studies reported that upregulation of Ezrin is a negative prognostic factor in various cancers. However, there is an opposite data indicating the involvement of negative or reduced expression of Ezrin in cancer progression [96]. This contradiction can be explained by the fact that Ezrin is implicated in the regulation not only of cell motility but also of cell adhesion, ion channels, cell proliferation, etc. [150].
The WAVE2 protein is involved in actin filament reorganization and lamellipodia formation and was shown to colocalize with Arp2 at the invasive front of breast cancer [97,147].
Cortactin regulates cortical actin cytoskeleton dynamics by stabilizing F-actin networks and promoting actin polymerization via activating the Arp2/3 complex [47,151]. According to in vitro and in vivo experiments, cortactin promotes invasion of head and neck tumors [151], and its high expression is found in the invasive front of oral and laryngeal tumors [98,99].
The MENA protein regulates actin polymerization and cell migration. An elevated level of the MENAinv isoform, which is involved in the formation of invadopodia due to phosphorylation of cortactin and activation of the N-WASP/Arp2/3 complex, is found in invasive cells of human tumors and animal tumor models and is associated with a high risk of metastasis [100,152,153].
Fascin-1 is an actin-binding protein involved in filopodia formation. It is highly expressed in nervous tissue and is normally absent in epithelial cells. However, a high level of fascin-1 is found in many malignant neoplasms of the liver, gallbladder, stomach, intestines, lung, breast, etc., and is a marker of poor prognosis [154,155]. Increased expression of fascin-1 is found in the invasive front of liver, colon, cervical, and endometrial cancers and is associated with a high risk of metastasis [101,102,103,104].

2.6. Other Proteins

In the invasive front, there are highly proliferating tumor cells, which probably facilitate the more efficient dissemination of the tumor. Expression of Ki-67, a cell proliferation marker, was shown to be elevated in the invasive front of oral and endometrial cancers [6,105,106]. In breast cancer, nuclear expression of Ki-67 is two-fold higher in the invasive front than in other parts of the tumor and is associated with metastasis to bones and liver [107]. Increased proliferation of tumor cells in the invasive front is also indicated by elevated expression of FGFR2 that is involved in the induction of signaling pathways affecting division, growth, and differentiation of cells, as demonstrated in colorectal and cervical cancers [109,110]. However, there are also contradictory data on negative expression Ki-67 or the absence of differences in its level between the invasive front and the tumor center in oral and colorectal cancers [56,69,108]. Moreover, FGFR2 is a multifunctional protein that regulates different biological processes such as proliferation, differentiation, etc. [111].
At first glance, the prevalence of cell proliferation in the invasive front is in contradiction to the data that invading tumor cells are enriched in EMT markers [63] because EMT typically associates with cell cycle arrest [156]. However, in the invasive front, EMT-cell cycle connection can be broken. In other words, instead of “go-or-grow”, tumor cells follow “go-and-grow” behavior [115,157].
The search for tumor invasion markers is an important issue aimed at assessing the risk of cancer metastasis. The role of the discussed molecules as invasive markers is controversial in most cases. Most of these molecules are involved not only in invasive growth but also in processes not related to cell migration. Nevertheless, some molecules such as WAVE2, cortactin, MENAinv, and fascin-1 are promising candidates for future studies of their roles as cancer cell invasion markers. In any case, the search for more specific markers of invasive growth is needed. In this regard, we think that the emphasis on intratumor morphological heterogeneity typical of many cancers may be very productive. In particular, investigation of the molecular make-up of various invasive tumor structures may enable identification of new molecules associated with invasion of tumor cells.

3. Intratumor Morphological Heterogeneity as a Model for Studying Cancer Cell Invasion

Based on more than 10-year morphological studies and detailed analysis of various structural features of invasive carcinoma of no special type of the breast (IC NST, previously classified as invasive ductal carcinoma), we have concluded that there are two types of tumors: Nonstructural and structural (Figure 1). Nonstructural breast carcinomas are characterized by a monomorphic pattern and are represented by large solid areas connected to each other, with thin layers of stromal elements (Figure 1).
Structural tumors are characterized by a polymorphic pattern and a pronounced phenotypic variety of the infiltrative (invasive) and stromal components (Figure 1). In other words, structural tumors demonstrate significant morphological heterogeneity. In initial attempts to determine the potential morphological IC NST features associated with cancer progression, we identified five main types of the invasive component in the tumor: Tubular, alveolar, solid, and trabecular structures, and discrete groups of tumor cells [158,159,160,161]. The tubular structures are tube-shaped and lumen-containing arrangements of single rows of rather monomorphic tumor cells with round monomorphic nuclei. The alveolar structures are clusters of round or slightly irregular tumor cells of different sizes, often with polymorphic nuclei. The number of cells in alveolar structures varies from 5–20. The solid structures are represented by large masses differing in size and shape, which consist of either small tumor cells with moderate cytoplasm and monomorphic nuclei or large cells with abundant cytoplasm and polymorphic nuclei. Although solid groups of tumor cells are a characteristic feature of nonstructural breast tumors, they are also observed in structural carcinomas. The trabecular structures are represented by either a single row of tumor cells (≥5 cells) or arrangements consisting of two rows of closely related monomorphic cells with moderate cytoplasm, which are parallel to each other. The discrete groups consist of small cell clusters (up to five cells) and single tumor cells (Figure 1). The size and shape of these cells and nuclei vary significantly [158,159,160,161].
Different morphological structures were shown to represent transcriptionally distinct tumor cell populations differing in the number of CD44+CD24 cancer stem cells, epithelial and mesenchymal features, and enrichment of cancer invasion signaling pathways [160]. Tubular and alveolar structures are similar in gene expression and demonstrate co-expression of epithelial and mesenchymal markers. The solid structures retain the epithelial features but demonstrate an increase in the mesenchymal traits and collective cell migration hallmarks. Trabecular and discrete groups are enriched in mesenchymal genes and cancer invasion pathways. CD44+CD24 cells are less common in the discrete groups and more abundant in the alveolar and solid structures [160]. Taken together, these data suggest that different morphological structures demonstrate varying degrees of EMT: From low in tubular, alveolar, and solid structures to advanced in trabecular and discrete groups of tumor cells [160].
The intratumor morphological heterogeneity of breast cancer is not an occasional phenomenon and is strongly associated with disease prognosis and therapy efficacy. Breast tumors with either alveolar or trabecular structures are characterized by a high rate of lymph node metastasis [161,162]. In neoadjuvant chemotherapy (NAC), tumors with alveolar or trabecular structures often demonstrate a poor response [162,163] and an increased risk of distant metastasis [162]. NAC-treated patients with alveolar or trabecular structures in breast tumors have decreased metastasis-free survival [162].
In a longitudinal study of the morphological, molecular genetic and clinical features of breast cancer, we have clearly seen that the differences are present not only in the structural pattern of tumor tissue. It has become obvious that breast carcinoma is characterized by pronounced intratumor morphological heterogeneity when morphologically similar and almost identical structures can exhibit completely different expression profiles, and it may not be ruled out that this phenomenon may somehow affect the behavior of tumor [66]. This conclusion prompted us to differentiate in more detail the previously described morphological structures.
A morphological analysis of structural IC NSTs revealed significant diversity and variability in solid groups of tumor cells, among which we identified two different variants: Solid structures with large torpedo-like sprouts and solid structures with small bud-like sprouts (Figure 1). The first variant is represented by various differently-sized, merging solid areas of tightly packed tumor cells connected with each other. In these structures, there are elongated, mostly triangular sprouts consisting of two–three parallel cell rows. The base of torpedo-like sprouts is always pointed out to the body of solid structures, while the tip, consisting of one–three tumor cells, penetrates to different depths to the stroma. Importantly, torpedo-like sprouts can be presented as structures independent of solid groups of tumor cells (Figure 1). Another variant of solid structures is represented by the large masses of tumor cells. However, a distinctive feature is that any edge of a solid structure comprises rounded or spherical bud-like sprouts consisting of five–seven atypical cells penetrating to the stroma (Figure 1).
Thus, the structural diversity of the infiltrative component and the pronounced intratumor morphological heterogeneity in IC NST represent an attractive model for investigation of tumor cell invasion. The solid structures both with large torpedo-like and small bud-like sprouts, as well as trabecular structures, may be considered as a morphological manifestation of collective cell invasion. Discrete groups of tumor cells, mainly single tumor cells, are an example of individual cell invasion.

4. Conclusions

Invasion is a key event towards the acquisition of the metastatic phenotype by tumor cells and an attractive target for anticancer therapy aimed at the prevention of metastasis. In in vitro studies, EMT has been proved to play an important role in the appearance of migrating and invading tumor cells. However, the cell movement mechanisms working in vitro are frequently not related to the invasive growth in vivo. Molecules that have been identified in vitro to be involved in cancer cell invasion do not demonstrate selective expression at the invasive front or at the tips of invasive structures where tumor cells are rather motile. Moreover, the expression of these molecules does not often demonstrate clinical significance for the prediction of cancer metastasis risk. Thus, the question how to identify invading tumor cells in human cancer specimens remains unanswered. In this regard, new effective models should be developed to investigate the mechanisms of cancer cell invasion. In our opinion, one of these models, at least in case of breast cancer, can be intratumor morphological heterogeneity which is a manifestation of different patterns of tumor cell invasion. The investigation of the molecular make-up of invasive structures of tumor cells and their microenvironment may provide valuable information about new molecules involved in the invasive growth and may identify novel prognostic markers and therapeutic targets.

Author Contributions

Conceptualization, V.M.P. and M.V.Z.; writing—original draft preparation, T.S.G., N.M.N, N.V.K., and S.Y.Z.; writing—review and editing, E.V.D.; supervision, N.V.C.

Funding

The study was supported by the Russian Science Foundation (grant #19-75-30016).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Hanahan, D.; Weinberg, R.A. Review Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [PubMed]
  2. Jiang, W.G.; Sanders, A.J.; Katoh, M.; Ungefroren, H.; Gieseler, F.; Prince, M.; Thompson, S.K.; Zollo, M.; Spano, D.; Dhawan, P.; et al. Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin. Cancer Biol. 2015, 35, 244–275. [Google Scholar]
  3. Sahai, E. Mechanisms of cancer cell invasion. Curr. Opin. Genet. Dev. 2005, 15, 87–96. [Google Scholar] [PubMed]
  4. Bànkfalvi, A.; Piffkò, J. Prognostic and predictive factors in oral cancer: The role of the invasive tumour front. J. Oral Pathol. Med. 2000, 29, 291–298. [Google Scholar] [PubMed]
  5. Zlobec, I.; Lugli, A. Invasive front of colorectal cancer: Dynamic interface of pro-/anti-tumor factors. World J. Gastroenterol. 2009, 15, 5898–5906. [Google Scholar] [PubMed]
  6. Rivera, C.; Venegas, B. Histological and molecular aspects of oral squamous cell carcinoma. Oncol. Lett. 2014, 8, 7–11. [Google Scholar] [PubMed]
  7. Friedl, P.; Wolf, K. Tumour-cell invasion and migration: Diversity and escape mechanisms. Nat. Rev. Cancer 2003, 3, 362–374. [Google Scholar]
  8. Wolf, K.; Friedl, P. Molecular mechanisms of cancer cell invasion and plasticity. Br. J. Dermatol. 2006, 154, 11–15. [Google Scholar]
  9. Krakhmal, N.V.; Zavyalova, M.V.; Denisov, E.V.; Vtorushin, S.V.; Perelmuter, V.M. Cancer invasion: Patterns and mechanisms. Acta Naturae 2015, 7, 17–28. [Google Scholar]
  10. Lintz, M.; Muñoz, A.; Reinhart-King, C.A. The Mechanics of Single Cell and Collective Migration of Tumor Cells. J. Biomech. Eng. 2017, 139, 021005. [Google Scholar]
  11. Wang, X.; Enomoto, A.; Asai, N.; Kato, T.; Takahashi, M. Collective invasion of cancer: Perspectives from pathology and development. Pathol. Int. 2016, 66, 183–192. [Google Scholar] [PubMed] [Green Version]
  12. Pandya, P.; Orgaz, J.L.; Sanz-Moreno, V. Modes of invasion during tumour dissemination. Mol. Oncol. 2017, 11, 5–27. [Google Scholar] [PubMed]
  13. Friedl, P.; Alexander, S. Cancer invasion and the microenvironment: Plasticity and reciprocity. Cell 2011, 147, 992–1009. [Google Scholar] [PubMed]
  14. 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. 2012, 109, E2595–E2604. [Google Scholar] [PubMed]
  15. 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] [PubMed]
  16. Cheung, K.J.; Ewald, A.J. Illuminating breast cancer invasion: Diverse roles for cell-cell interactions. Curr. Opin. Cell Biol. 2014, 30, 99–111. [Google Scholar] [PubMed]
  17. Paňková, K.; Rösel, D.; Novotný, M.; Brábek, J. The molecular mechanisms of transition between mesenchymal and amoeboid invasiveness in tumor cells. Cell. Mol. Life Sci. 2010, 67, 63–71. [Google Scholar] [PubMed]
  18. Chikina, A.S.; Alexandrova, A.Y. The cellular mechanisms and regulation of metastasis formation. Mol. Biol. 2014, 48, 165–180. [Google Scholar]
  19. Paluch, E.K.; Raz, E. The role and regulation of blebs in cell migration. Curr. Opin. Cell Biol. 2013, 25, 582–590. [Google Scholar] [Green Version]
  20. Huang, B.; Lu, M.; Jolly, M.K.; Tsarfaty, I.; Onuchic, J.; Ben-Jacob, E. The three-way switch operation of Rac1/RhoA GTPase-based circuit controlling amoeboid-hybrid-mesenchymal transition. Sci. Rep. 2014, 4, 1–11. [Google Scholar]
  21. Kalluri, R.; Weinberg, R. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 2009, 119, 1420–1428. [Google Scholar] [PubMed] [Green Version]
  22. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial–mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 1–45. [Google Scholar]
  23. Bonde, A.K.; Tischler, V.; Kumar, S.; Soltermann, A.; Schwendener, R.A. Intratumoral macrophages contribute to epithelial-mesenchymal transition in solid tumors. BMC Cancer 2012, 12, 35. [Google Scholar]
  24. Nieto, M.A.; Huang, R.Y.Y.J.; Jackson, R.A.A.; Thiery, J.P.P. Emt: 2016. Cell 2016, 166, 21–45. [Google Scholar] [PubMed]
  25. Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [PubMed] [Green Version]
  26. Gao, D.; Vahdat, L.T.; Wong, S.; Chang, J.C.; Mittal, V. Microenvironmental regulation of epithelial-mesenchymal transitions in cancer. Cancer Res. 2012, 72, 4883–4889. [Google Scholar] [PubMed]
  27. Jolly, M.K.; Boareto, M.; Huang, B.; Jia, D.; Onuchic, J.N.; Levine, H. Implications of the hybrid epithelial/mesenchymal phenotype in metastasis. Front Oncol. 2015, 5, 1–19. [Google Scholar]
  28. Grigore, A.D.; Jolly, M.K.; Jia, D. Tumor Budding: The Name is EMT. Partial EMT. J. Clin. Med. 2016, 5, 1–23. [Google Scholar]
  29. Zhang, S.; Wang, X.; Gupta, A.; Fang, X.; Wang, L.; Zhang, C. Expression of IL-17 with tumor budding as a prognostic marker in oral squamous cell carcinoma. Am. J. Transl. Res. 2019, 11, 1876–1883. [Google Scholar]
  30. Lorenzo Soriano, L.; Ordaz Jurado, G.; Pontones Moreno, J.L.; Villarroya Castillo, S.; Hernández Girón, S.; Sáez Moreno, I.; Ramos Soler, D. Tumor Budding: Prognostic Value in Muscle-Invasive Bladder Cancer. Urology 2019. [Google Scholar]
  31. Sirin, A.H.; Sokmen, S.; Unlu, S.M.; Ellidokuz, H.; Sarioglu, S. The prognostic value of tumor budding in patients who had surgery for rectal cancer with and without neoadjuvant therapy. Tech. Coloproctol. 2019, 23, 333–342. [Google Scholar] [PubMed]
  32. Ekmekci, S.; Kucuk, U.; Kokkoz, S.; Cakir, E.; Gumussoy, M. Tumor budding in laryngeal carcinoma. Indian J. Pathol. Microbiol. 2019, 62, 7–10. [Google Scholar] [PubMed]
  33. Agarwal, R.; Khurana, N.; Singh, T.; Agarwal, P.N. Tumor budding in infiltrating breast carcinoma: Correlation with known clinicopathological parameters and hormone receptor status. Indian J. Pathol. Microbiol. 2019, 62, 222–225. [Google Scholar] [PubMed]
  34. Ogino, M.; Nakanishi, Y.; Mitsuhashi, T.; Hatanaka, Y.; Amano, T.; Marukawa, K.; Nitta, T.; Ueno, T.; Ono, M.; Kuwabara, S.; et al. Impact of Tumour Budding Grade in 310 Patients Who Underwent Surgical Resection for Extrahepatic Cholangiocarcinoma. Histopathology 2019, 74, 861–872. [Google Scholar] [PubMed]
  35. Huang, B.; Jolly, M.K.; Lu, M.; Tsarfaty, I.; Ben-Jacob, E.; Onuchic, J.N. Modeling the Transitions between Collective and Solitary Migration Phenotypes in Cancer Metastasis. Sci. Rep. 2015, 5, 1–13. [Google Scholar]
  36. Lehmann, S.; Boekhorst, V.; Odenthal, J.; Bianchi, R.; van Helvert, S.; Ikenberg, K.; Ilina, O.; Stoma, S.; Xandry, J.; Jiang, L.; et al. Hypoxia Induces a HIF-1-Dependent Transition from Collective-to-Amoeboid Dissemination in Epithelial Cancer Cells. Curr. Biol. 2017, 27, 392–400. [Google Scholar] [PubMed] [Green Version]
  37. Bockhorn, M.; Jain, R.K.; Munn, L.L. Active versus passive mechanisms in metastasis: do cancer cells crawl into vessels, or are they pushed? Lancet Oncol. 2007, 8, 444–448. [Google Scholar] [Green Version]
  38. Iguchi, T.; Aishima, S.; Taketomi, A.; Nishihara, Y.; Fujita, N.; Sanefuji, K.; Maehara, Y.; Tsuneyoshi, M. Extracapsular penetration is a new prognostic factor in human hepatocellular carcinoma. Am. J. Surg. Pathol. 2008, 32, 1675–1682. [Google Scholar] [PubMed]
  39. Mehes, G.; Witt, A.; Kubista, E.; Ambros, P.F. Circulating Breast Cancer Cells Are Frequently Apoptotic. Am. J. Pathol. 2001, 159, 17–20. [Google Scholar] [PubMed] [Green Version]
  40. Larson, C.J.; Moreno, J.G.; Pienta, K.J.; Gross, S.; Repollet, M.; O’Hara, S.M.; Russell, T.; Terstappen, L.W. Apoptosis of circulating tumor cells in prostate cancer patients. Cytom. Part A 2004, 62, 46–53. [Google Scholar]
  41. Hood, J.D.; Cheresh, D.A. Role of integrins in cell invasion and migration. Nat. Rev. 2002, 2, 1–10. [Google Scholar]
  42. Rowe, R.G.; Weiss, S.J. Navigating ECM Barriers at the Invasive Front: The Cancer Cell—Stroma Interface. Annu. Rev. Cell Dev. Biol. 2009, 25, 567–595. [Google Scholar] [PubMed]
  43. Sentani, K.; Matsuda, M. Clinicopathological significance of MMP-7, laminin γ2 and EGFR expression at the invasive front of gastric carcinoma. Gastric Cancer 2014, 17, 412–422. [Google Scholar] [PubMed]
  44. Halfter, W.; Oertle, P.; Monnier, C.A.; Camenzind, L.; Reyes-Lua, M.; Hu, H.; Candiello, J.; Labilloy, A.; Balasubramani, M.; Henrich, P.B.; et al. New concepts in basement membrane biology. FEBS J. 2015, 282, 4466–4479. [Google Scholar] [PubMed]
  45. Hintermann, E.; Quaranta, V. Epithelial cell motility on laminin-5: Regulation by matrix assembly, proteolysis, integrins and erbB receptors. Matrix Biol. 2004, 23, 75–85. [Google Scholar] [PubMed]
  46. Masuda, R.; Kijima, H.; Imamura, N.; Aruga, N.; Nakazato, K.; Oiwa, K.; Nakano, T.; Watanabe, H.; Ikoma, Y.; Tanaka, M.; et al. Laminin-5 gamma 2 chain expression is associated with tumor cell invasiveness and prognosis of lung squamous cell carcinoma. Biomed. Res. 2012, 33, 309–317. [Google Scholar] [PubMed]
  47. Sharma, M.; Sah, P.; Sharma, S.S.; Radhakrishnan, R. Molecular changes in invasive front of oral cancer. J. Oral Maxillofac. Pathol. 2013, 17, 240–247. [Google Scholar] [PubMed]
  48. Ramovs, V.; te Molder, L.; Sonnenberg, A. The opposing roles of laminin-binding integrins in cancer. Matrix Biol. 2017, 57, 213–243. [Google Scholar]
  49. Ishikawa, T.; Wondimu, Z.; Oikawa, Y.; Gentilcore, G.; Kiessling, R.; Egyhazi Brage, S.; Hansson, J.; Patarroyo, M. Laminins 411 and 421 differentially promote tumor cell migration via α6β1 integrin and MCAM (CD146). Matrix Biol. 2014, 38, 69–83. [Google Scholar]
  50. Wang, H.; Fu, W.; Im, J.H.; Zhou, Z.; Santoro, S.A.; Iyer, V.; DiPersio, C.M.; Yu, Q.C.; Quaranta, V.; Al-Mehdi, A.; et al. Tumor cell α3β1 integrin and vascular laminin-5 mediate pulmonary arrest and metastasis. J. Cell Biol. 2004, 164, 935–941. [Google Scholar]
  51. Maltseva, D.V.; Rodin, S.A. Laminins in Metastatic Cancer. Mol. Biol. 2018, 52, 350–371. [Google Scholar]
  52. Miyazaki, K. Laminin-5 (laminin-332): Unique biological activity and role in tumor growth and invasion. Cancer Sci. 2006, 97, 91–98. [Google Scholar] [PubMed]
  53. Yamamoto, H.; Kitadai, Y.; Yamamoto, H.; Oue, N.; Ohdan, H.; Yasui, W.; Kikuchi, A. Laminin γ2 Mediates Wnt5a-Induced Invasion of Gastric Cancer Cells. Gastroenterology 2009, 137, 242–252. [Google Scholar] [PubMed]
  54. Okada, K.-I.; Kijima, H.; Imaizumi, T.; Hirabayashi, K.; Matsuyama, M.; Yazawa, N.; Oida, Y.; Tobita, K.; Tanaka, M.; Dowaki, S.; et al. Stromal laminin-5γ2 chain expression is associated with the wall-invasion pattern of gallbladder adenocarcinoma. Biomed. Res. 2009, 30, 53–62. [Google Scholar] [PubMed]
  55. Niki, T.; Kohno, T.; Iba, S.; Moriya, Y.; Takahashi, Y.; Saito, M.; Maeshima, A. Frequent Co-Localization of Cox-2 and Laminin-5 γ2 Chain at the Invasive Front of Early-Stage Lung Adenocarcinomas. Am. J. Pathol. 2002, 160, 1129–1141. [Google Scholar] [PubMed]
  56. Karamitopoulou, E.; Zlobec, I.; Panayiotides, I.; Patsouris, E.S.; Peros, G.; Rallis, G.; Lapas, C.; Karakitsos, P.; Terracciano, L.M.; Lugli, A. Systematic analysis of proteins from different signaling pathways in the tumor center and the invasive front of colorectal cancer. Hum. Pathol. 2011, 42, 1888–1896. [Google Scholar] [PubMed]
  57. Lowy, C.M.; Oskarsson, T.; Lowy, C.M.; Oskarsson, T. Tenascin C in metastasis: A view from the invasive front. Cell Adh. Migr. 2015, 9, 112–124. [Google Scholar] [Green Version]
  58. García-Solano, J.; Conesa-Zamora, P.; Trujillo-Santos, J.; Torres-Moreno, D.; Mäkinen, M.J.; Pérez-Guillermo, M. Immunohistochemical expression profile of β-catenin, E-cadherin, P-cadherin, laminin-5γ2 chain, and SMAD4 in colorectal serrated adenocarcinoma. Hum. Pathol. 2012, 43, 1094–1102. [Google Scholar]
  59. Bae, Y.K.; Kim, A.; Kim, M.K.; Choi, J.E.; Kang, S.H.; Lee, S.J. Fibronectin expression in carcinoma cells correlates with tumor aggressiveness and poor clinical outcome in patients with invasive breast cancer. Hum. Pathol. 2013, 44, 2028–2037. [Google Scholar]
  60. Gopal, S.; Veracini, L.; Grall, D.; Butori, C.; Schaub, S.; Audebert, S.; Camoin, L.; Baudelet, E.; Adwanska, A.; Beghelli-De La Forest Divonne, S.; et al. Fibronectin-guided migration of carcinoma collectives. Nat. Commun. 2017, 8, 14105. [Google Scholar]
  61. De Oliveira Ramos, G.; Bernardi, L.; Lauxen, I.; Filho, M.S.A.; Horwitz, A.R.; Lamers, M.L. Fibronectin modulates cell adhesion and signaling to promote single cell migration of highly invasive oral squamous cell carcinoma. PLoS ONE 2016, 11, 1–18. [Google Scholar]
  62. Aishima, S.; Taguchi, K.; Terashi, T.; Matsuura, S.; Shimada, M.; Tsuneyoshi, M. Tenascin Expression at the Invasive Front Is Associated with Poor Prognosis in Intrahepatic Cholangiocarcinoma. Mod. Pathol. 2003, 16, 1019–1027. [Google Scholar] [PubMed] [Green Version]
  63. Gurzu, S. Epithelial-mesenchymal, mesenchymal-epithelial, and endothelial-mesenchymal transitions in malignant tumors: An update. World J. Clin. Cases 2015, 3, 393–404. [Google Scholar] [PubMed]
  64. Christofori, G. New signals from the invasive front. Nature 2006, 441, 444–450. [Google Scholar] [PubMed]
  65. Zhong, J.; Ogura, K.; Wang, Z.; Inuzuka, H. Degradation of the Transcription Factor Twist, an Oncoprotein that Promotes Cancer Metastasis. Discov Med. 2013, 15, 7–15. [Google Scholar] [PubMed]
  66. Krakhmal, N.V.; Zavyalova, M.V.; Perelmuter, V.M.; Vtorushin, S.V.; Slonimskaya, E.M.; Denisov, E.V. Heterogeneous expression of markers associated with invasive breast cancer (in Russ). Sib. J. Oncol. 2016, 15, 56–61. [Google Scholar]
  67. Brabletz, T.; Kalluri, R.; Nieto, M.A.; Weinberg, R.A. EMT in cancer. Nat. Rev. Cancer 2018, 18, 128–134. [Google Scholar]
  68. Brabletz, T.; Jung, A.; Reu, S.; Porzner, M.; Hlubek, F.; Kunz-Schughart, L.A.; Knuechel, R.; Kirchner, T. Variable β-catenin expression in colorectal cancers indicates tumor progression driven by the tumor environment. Proc. Natl. Acad. Sci. 2002, 98, 10356–10361. [Google Scholar]
  69. Pereira, C.H.; Morais, M.O.; Martins, A.F.L.; Soares, M.Q.S.; de C.G. Alencar, R.; Batista, A.C.; Leles, C.R.; Mendonça, E.F. Expression of adhesion proteins (E-cadherin and β-catenin) and cell proliferation (Ki-67) at the invasive tumor front in conventional oral squamous cell and basaloid squamous cell carcinomas. Arch. Oral Biol. 2016, 61, 8–15. [Google Scholar]
  70. Suzuki, H.; Masuda, N.; Shimura, T.; Araki, K.; Kobayashi, T.; Tsutsumi, S.; Asao, T.; Kuwano, H. Nuclear β-Catenin Expression at the Invasive Front and in the Vessels Predicts Liver Metastasis in Colorectal Carcinoma. Anticancer Res. 2008, 28, 1821–1830. [Google Scholar]
  71. Wang, L.; Cheng, H.; Liu, Y.; Wang, L.; Yu, W.; Zhang, G.; Chen, B.; Yu, Z.; Hu, S. Prognostic value of nuclear β-catenin overexpression at invasive front in colorectal cancer for synchronous liver metastasis. Ann. Surg. Oncol. 2011, 18, 1553–1559. [Google Scholar] [PubMed]
  72. Shamir, E.R.; Ewald, A.J. Adhesion in mammary development: Novel roles for E-cadherin in individual and collective cell migration. Curr. Top. Dev. Biol. 2015, 112, 353–382. [Google Scholar] [PubMed]
  73. Yang, G.Y.; Guo, S.; Dong, C.Y.; Wang, X.Q.; Hu, B.Y.; Liu, Y.F.; Chen, Y.W.; Niu, J.; Dong, J.H. Integrin αvβ6 sustains and promotes tumor invasive growth in colon cancer progression. World J. Gastroenterol. 2015, 21, 7457–7467. [Google Scholar] [PubMed]
  74. Stewart, R.L.; West, D.; Wang, C.; Weiss, H.L.; Gal, T.; Durbin, E.B.; O’Connor, W.; Chen, M.; O’Connor, K.L. Elevated integrin α6β4 expression is associated with venous invasion and decreased overall survival in non-small cell lung cancer. Hum. Pathol. 2016, 54, 174–183. [Google Scholar] [PubMed]
  75. Albelda, S.M.; Mette, S.A.; Elder, D.E.; Stewart, R.M.; Damjanovich, L.; Herlyn, M.; Buck, C.A. Integrin Distribution in Malignant Melanoma: Association of the β3 Subunit with Tumor Progression. Cancer Res. 1990, 50, 6757–6764. [Google Scholar] [PubMed]
  76. Koopman Van Aarsen, L.A.; Leone, D.R.; Ho, S.; Dolinski, B.M.; McCoon, P.E.; LePage, D.J.; Kelly, R.; Heaney, G.; Rayhorn, P.; Reid, C.; et al. Antibody-mediated blockade of integrin αvβ6 inhibits tumor progression in vivo by a transforming growth factor-β-regulated mechanism. Cancer Res. 2008, 68, 561–570. [Google Scholar]
  77. Chou, F.C.; Chen, H.Y.; Kuo, C.C.; Sytwu, H.K. Role of galectins in tumors and in clinical immunotherapy. Int. J. Mol. Sci. 2018, 19, 430. [Google Scholar]
  78. Richard, O. Hynes. Integrins: Bidirectional, Allosteric Signaling Machines. Cell 2002, 110, 673–687. [Google Scholar]
  79. Wu, M.-H.; Hong, H.-C.; Cheng, H.-W.; Pan, S.-H.; Liang, Y.-R.; Hong, T.-M.; Chiang, W.-F.; Wong, T.-Y.; Shieh, D.-B.; Shiau, A.-L.; et al. Galectin-1-Mediated Tumor Invasion and Metastasis, Up-Regulated Matrix Metalloproteinase Expression, and Reorganized Actin Cytoskeletons. Mol. Cancer Res. 2009, 7, 311–318. [Google Scholar]
  80. Toussaint, L.G.; Nilson, A.E.; Goble, J.M.; Ballman, K.V.; James, C.D.; Lefranc, F.; Kiss, R.; Uhm, J.H. Galectin-1, a gene preferentially expressed at the tumor margin, promotes glioblastoma cell invasion. Mol. Cancer 2012, 11, 32. [Google Scholar] [PubMed]
  81. Gavert, N.; Conacci-sorrell, M.; Gast, D.; Schneider, A.; Altevogt, P.; Brabletz, T.; Ben-Ze’ev, A. L1, a novel target of β-catenin signaling, transforms cells and is expressed at the invasive front of colon cancers. J. Cell Biol. 2005, 168, 633–642. [Google Scholar] [PubMed]
  82. Tsutsumi, S.; Morohashi, S.; Kudo, Y.; Akasaka, H.; Ogasawara, H.; Ono, M.; Takasugi, K.; Ishido, K.; Hakamada, K.; Kijima, H. L1 Cell adhesion molecule (L1CAM) expression at the cancer invasive front is a novel prognostic marker of pancreatic ductal adenocarcinoma. J. Surg. Oncol. 2011, 103, 669–673. [Google Scholar] [PubMed]
  83. Altevogt, P.; Doberstein, K.; Fogel, M. L1CAM in human cancer. Int. J. Cancer 2016, 138, 1565–1576. [Google Scholar]
  84. Shi, Z.; Stack, M.S. Urinary-type plasminogen activator (uPA) and its receptor (uPAR) in squamous cell carcinoma of the oral cavity. Biochem. J. 2007, 407, 153–159. [Google Scholar] [PubMed] [Green Version]
  85. Rømer, J.; Pyke, C.; Lund, L.R.; Ralfkiær, E.; Danø, K. Cancer cell expression of urokinase-type plasminogen activator receptor mRNA in squamous cell carcinomas of the skin. J. Invest. Dermatol. 2001, 116, 353–358. [Google Scholar] [PubMed]
  86. Mahmood, N.; Mihalcioiu, C.; Rabbani, S.A. Multifaceted Role of the Urokinase-Type Plasminogen Activator (uPA) and Its Receptor (uPAR): Diagnostic, Prognostic, and Therapeutic Applications. Front. Oncol. 2018, 8, 24. [Google Scholar] [PubMed] [Green Version]
  87. Kessenbrock, K.; Plaks, V.; Werb, Z. Matrix Metalloproteinases: Regulators of the Tumor Microenvironment. Cell 2010, 141, 52–67. [Google Scholar] [PubMed] [Green Version]
  88. Ondruschka, C.; Buhtz, P.; Motsch, C.; Freigang, B.; Schneider-Stock, R.; Roessner, A.; Boltze, C. Prognostic value of MMP-2, -9 and TIMP-1,-2 immunoreactive protein at the invasive front in advanced head and neck squamous cell carcinomas. Pathol. Res. Pract. 2002, 198, 509–515. [Google Scholar] [PubMed]
  89. Ntayi, C.; Labrousse, A.L.; Debret, R.; Birembaut, P.; Bellon, G.; Antonicelli, F.; Hornebeck, W.; Bernard, P. Elastin-Derived Peptides Upregulate Matrix Metalloproteinase-2-ediated Melanoma Cell Invasion Through Elastin-Binding Protein. J. Invest. Dermatol. 2004, 122, 256–265. [Google Scholar]
  90. Liu, X.P.; Kawauchi, S.; Oga, A.; Tsushimi, K.; Tsushimi, M. Prognostic significance of matrix metalloproteinase-7 (MMP-7) expression at the invasive front in gastric carcinoma. Jpn. J. Cancer Res. 2002, 7, 291–295. [Google Scholar]
  91. Kitoh, T.; Yanai, H.; Saitoh, Y.; Nakamura, Y.; Matsubara, Y.; Kitoh, H.; Yoshida, T.; Okita, K. Increased expression of matrix metalloproteinase-7 in invasive early gastric cancer. J. Gastroenterol. 2004, 39, 434–440. [Google Scholar] [PubMed]
  92. Adachi, Y.; Yamamoto, H.; Itoh, F.; Hinoda, Y.; Okada, Y.; Imai, K. Contribution of matrilysin (MMP-7) to the metastatic pathway of human colorectal cancers. Gut 1999, 45, 252–258. [Google Scholar] [PubMed]
  93. Planagumà, J.; Liljeström, M.; Alameda, F.; Bützow, R.; Virtanen, I.; Reventós, J.; Hukkanen, M. Matrix metalloproteinase-2 and matrix metalloproteinase-9 codistribute with transcription factors RUNX1 / AML1 and ETV5 / ERM at the invasive front of endometrial and ovarian carcinoma. Hum. Pathol. 2011, 42, 57–67. [Google Scholar] [PubMed]
  94. Sterz, C.M.; Kulle, C.; Dakic, B.; Makarova, G.; Böttcher, M.C.; Bette, M.; Werner, J.A.; Mandic, R. A basal-cell-like compartment in head and neck squamous cell carcinomas represents the invasive front of the tumor and is expressing MMP-9. Oral Oncol. 2010, 46, 116–122. [Google Scholar] [PubMed]
  95. Li, Q.; Gao, H.; Xu, H.; Wang, X.; Pan, Y.; Hao, F.; Qiu, X.; Stoecker, M.; Wang, E.; Wang, E. Expression of ezrin correlates with malignant phenotype of lung cancer, and in vitro knockdown of ezrin reverses the aggressive biological behavior of lung cancer cells. Tumour Biol. 2012, 33, 1493–1504. [Google Scholar] [PubMed]
  96. Li, J.; Wei, K.; Yu, H.; Jin, D.; Wang, G.; Yu, B. Prognostic Value of Ezrin in Various Cancers: A Systematic Review and Updated Meta-analysis. Sci. Rep. 2015, 5, 1–13. [Google Scholar]
  97. Iwaya, K.; Norio, K.; Mukai, K. Coexpression of Arp2 and WAVE2 predicts poor outcome in invasive breast carcinoma. Mod. Pathol. 2007, 20, 339–343. [Google Scholar] [PubMed]
  98. Ambrosio, P.E.; Rosa, E.F.; Aparecida, M.; Domingues, C.; André, R.; Villacis, R.; Coudry, R.D.A.; Tagliarini, J.V.; Soares, F.A. Cortactin is associated with perineural invasion in the deep invasive front area of laryngeal carcinomas. Hum. Pathol. 2011, 42, 1221–1229. [Google Scholar] [PubMed]
  99. Yamada, S.I.; Yanamoto, S.; Kawasaki, G.; Mizuno, A.; Nemoto, T.K. Overexpression of cortactin increases invasion potential in oral squamous cell carcinoma. Pathol. Oncol. Res. 2010, 16, 523–531. [Google Scholar] [PubMed]
  100. Toyoda, A.; Yokota, A.Y.A.; Saito, T.; Kawana, H. Overexpression of human ortholog of mammalian enabled (hMena) is associated with the expression of mutant p53 protein in human breast cancers. Int. J. Oncol. 2011, 38, 89–96. [Google Scholar]
  101. Vignjevic, D.; Schoumacher, M.; Gavert, N.; Janssen, K.; Jih, G.; Lae, M.; Louvard, D.; Ben-ze, A.; Robine, S. Fascin, a Novel Target of β-Catenin-TCF Signaling, Is Expressed at the Invasive Front of Human Colon Cancer. Cancer Res. 2007, 67, 6844–6854. [Google Scholar] [PubMed]
  102. Won, K.Y.; Kim, G.Y.; Lim, S.J.; Park, Y.K.; Kim, Y.W. Prognostic significance of fascin expression in extrahepatic bile duct carcinomas. Pathol. Res. Pract. 2009, 205, 742–748. [Google Scholar] [PubMed]
  103. Stewart, C.J.R.; Crook, M.; Loi, S. Fascin expression in endocervical neoplasia: Correlation with tumour morphology and growth pattern. J. Clin. Pathol. 2012, 65, 213–217. [Google Scholar] [PubMed]
  104. Stewart, C.J.R.; Crook, M.L. Fascin expression in undifferentiated and dedifferentiated endometrial carcinoma. Hum. Pathol. 2015, 46, 1514–1520. [Google Scholar] [PubMed]
  105. Kurokawa, H.; Zhang, M.; Matsumoto, S.; Yamashita, Y.; Tanaka, T.; Tomoyose, T.; Takano, H.; Funaki, K.; Fukuyama, H.; Takahashi, T.; et al. The relationship of the histologic grade at the deep invasive front and the expression of Ki-67 antigen and p53 protein in oral squamous cell carcinoma. J. Oral Pathol. Med. 2005, 34, 602–607. [Google Scholar] [PubMed]
  106. Horre, N.; Van Diest, P.J.; Sie-Go, D.M.; Heintz, A.P. The invasive front in endometrial carcinoma: higher proliferation and associated derailment of cell cycle regulators. Hum. Pathol. 2007, 38, 1232–1238. [Google Scholar]
  107. Gong, P.; Wang, Y.; Liu, G.; Zhang, J.; Wang, Z. New Insight into Ki67 Expression at the Invasive Front in Breast Cancer. PLoS ONE 2013, 8, 1–5. [Google Scholar]
  108. Rubio, C.A. Further studies on the arrest of cell proliferation in tumor cells at the invading front of colonic adenocarcinoma. J. Gastroenterol. Hepatol. 2007, 22, 1877–1881. [Google Scholar]
  109. Matsuda, Y.; Ishiwata, T.; Yamahatsu, K.; Kawahara, K. Overexpressed fibroblast growth factor receptor 2 in the invasive front of colorectal cancer: A potential therapeutic target in colorectal cancer. Cancer Lett. 2011, 309, 209–219. [Google Scholar]
  110. Kawase, R.; Ishiwata, T.; Matsuda, Y.; Onda, M.; Kudo, M.; Takeshita, T.; Naito, Z. Expression of fibroblast growth factor receptor 2 IIIc in human uterine cervical intraepithelial neoplasia and cervical cancer. Int. J. Oncol. 2010, 38, 257–266. [Google Scholar]
  111. Turner, N.; Grose, R. Fibroblast growth factor signalling: From development to cancer. Nat. Rev. Cancer 2010, 10, 116–129. [Google Scholar] [PubMed]
  112. Pal, S.; Ganguly, K.K.; Chatterjee, A. Extracellular matrix protein fibronectin induces matrix metalloproteinases in human prostate adenocarcinoma cells PC-3. Cell Commun. Adhes. 2013, 20, 105–114. [Google Scholar] [PubMed]
  113. Wang, J.P.; Hielscher, A. Fibronectin: How its aberrant expression in tumors may improve therapeutic targeting. J. Cancer 2017, 8, 674–682. [Google Scholar] [PubMed]
  114. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.; Lebleu, V.S.; Kalluri, R.; Biology, C. EMT Program is Dispensable for Metastasis but Induces Chemoresistance in Pancreatic Cancer. Nature 2016, 527, 525–530. [Google Scholar]
  115. Jolly, M.K.; Ware, K.E.; Gilja, S.; Somarelli, J.A.; Levine, H. EMT and MET: necessary or permissive for metastasis? Mol. Oncol. 2017, 11, 755–769. [Google Scholar] [PubMed]
  116. Savagner, P. Epithelial-mesenchymal transitions: From cell plasticity to concept elasticity. Curr. Top Dev. Biol. 2015, 112, 273–300. [Google Scholar] [PubMed]
  117. Shamir, E.R.; Pappalardo, E.; Jorgens, D.M.; Coutinho, K.; Tsai, W.T.; Aziz, K.; Auer, M.; Tran, P.T.; Bader, J.S.; Ewald, A.J. Twist1-induced dissemination preserves epithelial identity and requires E-cadherin. J. Cell Biol. 2014, 204, 839–856. [Google Scholar]
  118. Isaeva, A.V.; Zima, A.P.; Shabalova, I.P.; Ryazantseva, N.V.; Vasil’eva, O.A.; Kasoayn, K.T.; Saprina, T.V.; Latypova, V.N.; Berezkina, I.S.; Novitskii, V.V. β-catenin: Structure, function and role in malignant transformation of epithelial cells (in Russ). Vestn. Ross. Akad. Meditsinskikh Nauk 2015, 70, 475–483. [Google Scholar]
  119. Jolly, M.K.; Boareto, M.; Debeb, B.G.; Aceto, N.; Farach-Carson, M.C.; Woodward, W.A.; Levine, H. Inflammatory breast cancer: A model for investigating cluster-based dissemination. NPJ Breast Cancer 2017, 3, 1–7. [Google Scholar]
  120. Friedl, P.; Gilmour, D. Collective cell migration in morphogenesis, regeneration and cancer. Nat. Rev. 2009, 10, 455–456. [Google Scholar]
  121. Rathinam, R.; Alahari, S.K. Important role of integrins in the cancer biology. Cancer Metastasis Rev. 2010, 29, 223–237. [Google Scholar] [PubMed]
  122. Huttenlocher, A.; Horwitz, A.R. Integrins in Cell Migration. Cold Spring Harb. Perspect. Biol. 2011, 1–17. [Google Scholar]
  123. Ganguly, K.K.; Pal, S.; Moulik, S.; Chatterjee, A. Integrins and metastasis. Cell Adhes. Migr. 2013, 7, 251–261. [Google Scholar] [Green Version]
  124. Morini, M.; Marcella, M.; Nicoletta, F.; Federica, G.; Simonetta, B.; Roberta, M.; Douglas, M.N.; Pier Giorgio, N.; Adriana, A. The α3β1 integrin is associated with mammary carcinoma cell metastasis, invasion, and gelatinase B (mmp-9) activity. Int. J. Cancer 2000, 87, 336–342. [Google Scholar] [PubMed]
  125. He, Y.; Liu, X.D.; Chen, Z.Y.; Zhu, J.; Xiong, Y.; Li, K.; Dong, J.H.; Li, X. Interaction between cancer cells and stromal fibroblasts is required for activation of the uPAR-uPA-MMP-2 cascade in pancreatic cancer metastasis. Clin. Cancer Res. 2007, 13, 3115–3124. [Google Scholar] [PubMed]
  126. 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]
  127. Guo, W.; Giancotti, F.G. Integrin signalling during tumour progression. Nat. Rev. Mol. Cell Biol. 2004, 5, 816–826. [Google Scholar] [PubMed]
  128. Chan, B.M.; Matsuura, N.; Takada, Y.; Zetter, B.R.; Hemler, M.E. In vitro and in vivo consequences of VLA-2 expression on rhabdomyosarcoma cells. Science 1991, 251, 1600–1602. [Google Scholar]
  129. Desgrosellier, J.S.; Cheresh, D.A. Integrins in cancer: Biological implications and therapeutic opportunities. Nat. Rev. Cancer 2010, 10, 9–22. [Google Scholar]
  130. Wu, L.; Bernard-Trifilo, J.A.; Lim, Y.; Lim, S.T.; Mitra, S.K.; Uryu, S.; Chen, M.; Pallen, C.J.; Cheung, N.K.V.; Mikolon, D.; et al. Distinct FAK-Src activation events promote α5β1 and α4β1 integrin-stimulated neuroblastoma cell motility. Oncogene 2008, 27, 1439–1448. [Google Scholar]
  131. Schöttelndreier, H.; Potter, B.V.; Mayr, G.W.; Guse, A.H. Mechanisms involved in α6β1-integrin-mediated Ca(2+) signalling. Cell. Signal. 2001, 13, 895–899. [Google Scholar] [PubMed]
  132. Inoue, O.; Suzuki-Inoe, K.; McCarty, O.J.T.; Moroi, M.; Ruggeri, Z.M.; Kunicki, T.J. Laminin stimulates spreading of platelets through integrin α6β1–dependent activation of GPVI. Northwest Sci. 2006, 107, 1405–1411. [Google Scholar]
  133. Rapoport, E.M.; Kurmyshkina, O.V.; Bovin, N.V. Mammalian galectins: Structure, carbohydrate specificity, and functions. Biochem. 2008, 73, 393–405. [Google Scholar]
  134. Kowitz, A.; Kadmon, G.; Verschueren, H.; Remels, L.; De Baetselier, P.; Hubbe, M.; Schachner, M.; Schirrmacher, V.; Altevogt, P. Expression of L1 cell adhesion molecule is associated with lymphoma growth and metastasis. Clin. Exp. Metastasis 1993, 11, 419–429. [Google Scholar] [PubMed]
  135. Sidenius, N.; Blasi, F. The urokinase plasminogen activator system in cancer: Recent advances and implication for prognosis and therapy. Cancer Metastasis Rev. 2003, 22, 205–222. [Google Scholar] [PubMed]
  136. Banys-Paluchowski, M.; Witzel, I.; Aktas, B.; Fasching, P.A.; Hartkopf, A.; Janni, W.; Kasimir-Bauer, S.; Pantel, K.; Schön, G.; Rack, B.; et al. The prognostic relevance of urokinase-type plasminogen activator (uPA) in the blood of patients with metastatic breast cancer. Sci. Rep. 2019, 9, 1–10. [Google Scholar]
  137. Pavón, M.A.; Arroyo-Solera, I.; Céspedes, M.V.; Casanova, I.; León, X.; Mangues, R. uPA/uPAR and SERPINE1 in head and neck cancer: Role in tumor resistance, metastasis, prognosis and therapy. Oncotarget 2016, 7, 57351–57366. [Google Scholar]
  138. Bozzuto, G.; Ruggieri, P.; Molinari, A. Molecular aspects of tumor cell migration and invasion. Ann Ist Super Sanita 2010, 46, 66–80. [Google Scholar]
  139. Mohtasham, N.; Babakoohi, S.; Shiva, A.; Shadman, A.; Kamyab-Hesari, K.; Shakeri, M.-T.; Sharifi-Sistan, N. Immunohistochemical study of p53, Ki-67, MMP-2 and MMP-9 expression at invasive front of squamous cell and verrucous carcinoma in oral cavity. Pathol. Res. Pract. 2013, 209, 110–114. [Google Scholar]
  140. Mu, D.; Cambier, S.; Fjellbirkeland, L.; Baron, J.L.; Munger, J.S.; Kawakatsu, H.; Sheppard, D.; Courtney Broaddus, V.; Nishimura, S.L. The integrin ανβ8 mediates epithelial homeostasis through MT1-MMP-dependent activation of TGF-β1. J. Cell Biol. 2002, 157, 493–507. [Google Scholar]
  141. Yu, Q.; Stamenkovic, I. Cell surface-localized matrix metalloproteinase-9 proteolytically activates TGF-β and promotes tumor invasion and angiogenesis. Genes Dev. 2000, 14, 163–176. [Google Scholar] [PubMed]
  142. Tatti, O.; Vehviläinen, P.; Lehti, K.; Keski-Oja, J. MT1-MMP releases latent TGF-β1 from endothelial cell extracellular matrix via proteolytic processing of LTBP-1. Exp. Cell Res. 2008, 314, 2501–2514. [Google Scholar] [PubMed]
  143. Mitsiades, N.; Yu, W.H.; Poulaki, V.; Tsokos, M.; Stamenkovic, I. Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity. Cancer Res. 2001, 61, 577–581. [Google Scholar] [PubMed]
  144. Bergers, G.; Brekken, R.; McMahon, G.; Vu, T.H.; Itoh, T.; Tamaki, K.; Tanzawa, K.; Thorpe, P.; Itohara, S.; Werb, Z.; et al. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2000, 2, 737–744. [Google Scholar] [PubMed]
  145. Ahn, G.O.; Brown, J.M. Matrix metalloproteinase-9 is required for tumor vasculogenesis but not for angiogenesis: role of bone marrow-derived myelomonocytic cells. Cancer Cell 2008, 13, 193–205. [Google Scholar] [PubMed]
  146. Ardi, V.C.; Kupriyanova, T.A.; Deryugina, E.I.; Quigley, J.P. Human neutrophils uniquely release TIMP-free MMP-9 to provide a potent catalytic stimulator of angiogenesis. Proc. Natl. Acad. Sci. 2007, 104, 20262–20267. [Google Scholar] [PubMed] [Green Version]
  147. Yamaguchi, H.; Condeelis, J. Regulation of the actin cytoskeleton in cancer cell migration and invasion. Biochim. Biophys. Acta 2007, 642–652. [Google Scholar]
  148. Clucas, J.; Valderrama, F. ERM proteins in cancer progression. J. Cell Sci. 2015, 128, 1253. [Google Scholar] [PubMed]
  149. Martín-Villar, E.; Scholl, F.G.; Gamallo, C.; Yurrita, M.M.; Muñoz-Guerra, M.; Cruces, J.; Quintanilla, M. Characterization of human PA2.26 antigen (T1α-2, podoplanin), a small membrane mucin induced in oral squamous cell carcinomas. Int. J. Cancer 2005, 113, 899–910. [Google Scholar] [PubMed]
  150. Fehon, R.G.; McClatchey, A.I.; Bretscher, A. Organizing the cell cortex: The role of ERM proteins. Nat. Rev. Mol. Cell Biol. 2010, 11, 276–287. [Google Scholar] [PubMed]
  151. Clark, E.S.; Brown, B.; Whigham, A.S.; Kochaishvili, A.; Yarbrough, W.G.; Weaver, A.M. Aggressiveness of HNSCC tumors depends on expression levels of cortactin, a gene in the 11q13 amplicon. Oncogene 2009, 28, 431–444. [Google Scholar] [PubMed]
  152. Roussos, E.T.; Balsamo, M.; Alford, S.K.; Wyckoff, J.B.; Gligorijevic, B.; Wang, Y.; Pozzuto, M.; Stobezki, R.; Goswami, S.; Segall, J.E.; et al. Mena invasive (MenaINV) promotes multicellular streaming motility and transendothelial migration in a mouse model of breast cancer. J. Cell Sci. 2011, 124, 2120–2131. [Google Scholar] [PubMed] [Green Version]
  153. Weidmann, M.D.; Surve, C.R.; Eddy, R.J.; Chen, X.; Gertler, F.B.; Sharma, V.P.; Condeelis, J.S. MenaINV dysregulates cortactin phosphorylation to promote invadopodium maturation. Sci. Rep. 2016, 6, 1–18. [Google Scholar]
  154. Machesky, L.M.; Li, A. Fascin. Commun. Integr. Biol. 2010, 3, 263–270. [Google Scholar] [PubMed]
  155. Papaspyrou, K.; Brochhausen, C.; Schmidtmann, I.; Fruth, K.; Gouveris, H.; Kirckpatrick, J.; Mann, W.; Brieger, J. Fascin upregulation in primary head and neck squamous cell carcinoma is associated with lymphatic metastasis. Oncol. Lett. 2014, 7, 2041–2046. [Google Scholar] [PubMed]
  156. Lovisa, S.; LeBleu, V.S.; Tampe, B.; Sugimoto, H.; Vadnagara, K.; Carstens, J.L.; Wu, C.C.; Hagos, Y.; Burckhardt, B.C.; Pentcheva-Hoang, T.; et al. Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in renal fibrosis. Nat. Med. 2015, 21, 998–1009. [Google Scholar] [PubMed]
  157. Bauer, A.L.; Jackson, T.L.; Jiang, Y.; Rohlf, T. Receptor cross-talk in angiogenesis: Mapping environmental cues to cell phenotype using a stochastic, Boolean signaling network model. J. Theor. Biol. 2010, 264, 838–846. [Google Scholar] [PubMed]
  158. Zavyalova, M.V.; Perelmuter, V.M.; Slonimskaya, E.M.; Vtorushin, S.V.; Garbukov, E.Y.; Gluschenko, S.A. Conjugation of lymphogenous metastatic spread and histologic pattern of infiltrative component of ductal breast cancer (in Russ). Sib. J. Oncol. 2006, 1, 32–35. [Google Scholar]
  159. Perelmuter, V.M.; Zavyalova, M.V.; Slonimskaya, E.M.; Vtorushin, S.V.; Garbukov, E.Y. Hematogenous metastasis depending on histologic tumor pattern in breast cancer (In Russ). Sib. J. Oncol. 2006, 3, 29–33. [Google Scholar]
  160. Denisov, E.V.; Skryabin, N.A.; Gerashchenko, T.S.; Tashireva, L.A.; Wilhelm, J.; Buldakov, M.A.; Sleptcov, A.A.; Lebedev, I.N.; Vtorushin, S.V.; Zavyalova, M.V.; et al. Clinically relevant morphological structures in breast cancer represent transcriptionally distinct tumor cell populations with varied degrees of epithelial-mesenchymal transition and CD44+CD24 stemness. Oncotarget 2017, 8, 61163–61180. [Google Scholar]
  161. Zavyalova, M.V.; Perelmuter, V.M.; Vtorushin, S.V.; Denisov, E.V.; Litvyakov, N.V.; Slonimskaya, E.M.; Cherdyntseva, N.V. The Presence of Alveolar Structures in Invasive Ductal NOS Breast Carcinoma is Associated with Lymph Node Metastasis. Diagn. Cytopathol. 2013, 41, 279–282. [Google Scholar] [PubMed]
  162. Gerashchenko, T.S.; Zavyalova, M.V.; Denisov, E.V.; Krakhmal, N.V.; Pautova, D.N.; Litviakov, N.V.; Vtorushin, S.V.; Cherdyntseva, N.V.; Perelmuter, V.M. Intratumoral morphological heterogeneity of breast cancer as an indicator of the metastatic potential and tumor chemosensitivity. Acta Naturae 2017, 9, 56–67. [Google Scholar] [PubMed]
  163. Denisov, E.V.; Litviakov, N.V.; Zavyalova, M.V.; Perelmuter, V.M.; Vtorushin, S.V.; Tsyganov, M.M.; Gerashchenko, T.S.; Garbukov, E.Y.; Slonimskaya, E.M.; Cherdyntseva, N.V. Intratumoral morphological heterogeneity of breast cancer: neoadjuvant chemotherapy efficiency and multidrug resistance gene expression. Sci. Rep. 2014, 4, 1–7. [Google Scholar]
Figure 1. Two types of breast carcinomas based on a structural pattern. Nonstructural breast carcinomas are represented by large solid fields of cells connected to each other. Structural breast carcinomas are characterized by a phenotypic variety of the infiltrative (invasive) component, represented by certain types of morphological arrangements of tumor cells: Tubular structures, solid structures with small sprouts, solid structures with large torpedo-like sprouts, alveolar structures, torpedo-like structures, trabecular structures, and discrete groups of tumor cells. The images of hematoxylin and eosin-stained sections were obtained from the database of the Department of Pathological Anatomy, Siberian State Medical University, Tomsk, Russia.
Figure 1. Two types of breast carcinomas based on a structural pattern. Nonstructural breast carcinomas are represented by large solid fields of cells connected to each other. Structural breast carcinomas are characterized by a phenotypic variety of the infiltrative (invasive) component, represented by certain types of morphological arrangements of tumor cells: Tubular structures, solid structures with small sprouts, solid structures with large torpedo-like sprouts, alveolar structures, torpedo-like structures, trabecular structures, and discrete groups of tumor cells. The images of hematoxylin and eosin-stained sections were obtained from the database of the Department of Pathological Anatomy, Siberian State Medical University, Tomsk, Russia.
Jcm 08 01092 g001
Table 1. Potential markers of cancer cell invasion.
Table 1. Potential markers of cancer cell invasion.
MarkersFunctionsExpression at the Invasive FrontLimitations
ECM componentsLaminin-5, γ2 chainECM components, triggering MMP production through interaction with integrinsBreast, pancreatic, colon, lung, and other cancers [46,51,52,55,56]Expression not only in the invasive front, but in other regions of the tumor [43,46,57,58,59]
FibronectinOral and head and neck cancers [60,61]
Tenascin CModulation of cell adhesionMelanoma, breast, lung, liver, and gallbladder cancers [57,62]
EMT moleculesSnail, Twist, vimentinEMT induction and regulationVarious cancers [63]Snail and Twist: Unstable molecules [64,65], total expression in breast tumors [66]. Vimentin may not be expressed in invasive carcinomas [67]
Cell–cell and cell–ECM interaction moleculesCadherin-catenin complexAdherens junctionsColorectal, oral, and basaloid carcinomas (loss of E-cadherin and nuclear localization of β-catenin) [68,69,70,71]In some tumors, loss of E-cadherin is not indispensable for invasive growth [72]
IntegrinsCell–ECM adhesion, involvement in MMP productionMelanoma (αvβ3), colon (αvβ6), head and neck (αvβ6), and lung (α6β4) cancers [73,74,75,76]Involvement in other biological processes [77,78]
Galectin 1Modulation of cell–cell and cell–ECM interactionsOral and lung cancers, glioblastoma [47,79,80]
L1CAMCell adhesionColorectal and pancreatic cancers [81,82]Dualistic role in cancer progression [83]
Serine proteases and MMPsuPAProteolysis of plasminogen to plasminOral and skin carcinomas [84,85]Involvement in other biological processes [86,87]
MMPsECM proteolysisMelanoma (MMP-2), colorectal (MMP-7), gastric (MMP-7), endometrial (MMP-2, 9), ovarian (MMP-2, 9), and head and neck (MMP-2, 9) cancers [56,88,89,90,91,92,93,94]
Actin cytoskeleton proteinsEzrinActin polymerization, cytoskeletal dynamicsLung cancer [95,96]Involvement in other biological processes. Contradictory data on the role in cancer progression [96]
WAVE2Breast cancer [97]-
CortactinOral and laryngeal cancers [98,99]-
MENAinvBreast cancer [100]-
Fascin-1Liver, colon, cervical, and endometrial cancers [101,102,103,104]-
Other proteinsKi-67Cell proliferationBreast, oral, and endometrial cancers [6,105,106,107]Contradictory data on the level of Ki-67 expression at the invasive front [56,69,108]
FGFR2Cell division, growth and differentiationColorectal and cervical cancers [109,110]Involvement in other biological processes [111]
ECM, extracellular matrix; EMT, epithelial–mesenchymal transition; MMPs, matrix metalloproteinases.

Share and Cite

MDPI and ACS Style

Gerashchenko, T.S.; Novikov, N.M.; Krakhmal, N.V.; Zolotaryova, S.Y.; Zavyalova, M.V.; Cherdyntseva, N.V.; Denisov, E.V.; Perelmuter, V.M. Markers of Cancer Cell Invasion: Are They Good Enough? J. Clin. Med. 2019, 8, 1092. https://doi.org/10.3390/jcm8081092

AMA Style

Gerashchenko TS, Novikov NM, Krakhmal NV, Zolotaryova SY, Zavyalova MV, Cherdyntseva NV, Denisov EV, Perelmuter VM. Markers of Cancer Cell Invasion: Are They Good Enough? Journal of Clinical Medicine. 2019; 8(8):1092. https://doi.org/10.3390/jcm8081092

Chicago/Turabian Style

Gerashchenko, Tatiana S., Nikita M. Novikov, Nadezhda V. Krakhmal, Sofia Y. Zolotaryova, Marina V. Zavyalova, Nadezhda V. Cherdyntseva, Evgeny V. Denisov, and Vladimir M. Perelmuter. 2019. "Markers of Cancer Cell Invasion: Are They Good Enough?" Journal of Clinical Medicine 8, no. 8: 1092. https://doi.org/10.3390/jcm8081092

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