Tuesday, January 8, 2019

Epithelial to Mesenchymal Transitions and Cancer


When I was young and my father had returned from the Navy in WWII he had a phrase he used frequently to admonish my at times less than nest tendencies. Namely: "a place for everything and everything in its place". I thought I knew what he was saying but it was not until I started to understand cancer metastasis that this truly rang a bell. Cancer is not "neat". It just drops stuff all over the place, sending cells hither and thither, never putting things back where they belong.

We examine another process which is linked to cancer andmetastasis, namely the Epithelial to Mesenchymal Transition, EMT. As we have noted in many other areas we have examined this has been argued to have significant therapeutic interest. There has been some examination here as of later but there is limited clinical application. What this area does do is shine a light on the issue of cell location and lost of location stability as an integral part of cell carcinogenesis.

Cells express genes in different ways depending when and where they are. Epithelial cells generally express genes that allow the cell to perform a specific function and to do so at a specific location. However from time to time, such as in the growth phase of an organism, this stable phenotype is suppressed and the cell has a characteristic that allows it to move freely as a mesenchymal cell. Thus transitions from mesenchymal to epithelial phenotypes are stabilizing transitions in a maturing organism (called MET). The reverse, EMT, are generally destabilizing transitions. For example a melanocyte with E cadherin expressed binds to the other keratinocytes and remain stable in the skin. When E cadherin is not expressed but N cadherin is, the melanocytes bind together and then wander, often first upward creating a carcinoma in situ, then downward creating a melanoma. Thus as expression of genes is effected the process of EMT allows for movement and thus metastasis.

In this note, we examine some of the recent advances understanding this process, especially as applied to several somatic malignancies. There is also the consideration of using EMT mechanisms as a means to target therapeutics to mitigate metastasis. The state of the art is still somewhat early but it does provide an interesting alternative. This paper is not meant to be comprehensive but suggestive.

Weinberg presents a detailed description of the EMT as a part of metastasis[1].

As Heerboth et al note:

EMT and MET comprise the processes by which cells transit between epithelial and mesenchymal states, and they play integral roles in both normal development and cancer metastasis. This article reviews these processes and the molecular pathways that contribute to them.

First, we compare embryogenesis and development with cancer metastasis.

We then discuss the signaling pathways and the differential expression and down-regulation of receptors in both tumor cells and stromal cells, which play a role in EMT and metastasis.

We further delve into the clinical implications of EMT and MET in several types of tumors, and lastly, we discuss the role of epigenetic events that regulate EMT/MET processes.

We hypothesize that reversible epigenetic events regulate both EMT and MET, and thus, also regulate the development of different types of metastatic cancers.

The above is more of an outline of the issues that can be considered. Namely: (i) benign EMT processes versus malignant, (ii) downregulation and control of EMT pathways, (iii) specific EMT effects in specific cancers, and (iv) reversible controls regulating metastasis.

As Radisky notes:

The epithelial-mesenchymal transition (EMT) is an orchestrated series of events in which cell-cell and cell-extracellular matrix (ECM) interactions are altered to release epithelial cells from the surrounding tissue, the cytoskeleton is reorganized to confer the ability to move through a three-dimensional ECM, and a new transcriptional program is induced to maintain the mesenchymal phenotype.

Essential for embryonic development, EMT is nevertheless potentially destructive if deregulated, and it is becoming increasingly clear that inappropriate utilization of EMT mechanisms is an integral component of the progression of many tumors of epithelial tissues. Structural integrity is a key property of epithelial tissues: external epithelia serve as protective barriers against environmental hazards, and internal epithelia create defined and physiologically controlled subdomains within the organism. Epithelial structure is maintained by cell-cell interactions.

These involve tight junctions, cadherin based adherens junctions that are connected to the actin cytoskeleton, gap junctions that allow direct chemical interactions between neighboring cells, and desmosomes connected to the intermediate filament cytoskeleton, and cell-ECM interactions mediated by integrins and other molecules.

In this paper we address the following:

1. What is EMT and how does it function?
2. What are the critical drivers of the EMT process?
3. How does EMT effect a cancerous process?
4. What are the pathway elements involved in EMT?
5. What are the specifics of various cancers and EMT?
6. What is the interaction between the immune system and the EMT process?
7. What role does chronic inflammation play in EMT activation and in turn cancer?
8. What are the therapeutic opportunities available in the EMT context?
9. Does the presence of blood borne EMT markers present a diagnostic, prognostic, and therapeutic opportunity?


EMT is a process whereby a cell changes from a stable cell in a well defined matrix to a cell which has the ability to move about in a relatively unstructured manner. In essence the EMT process enables a metastatic change. We summarize some of these features herein.

EMT is simply the process whereby cells lose the ability to be at the right place at the right time. From Kalluri and Weinberg we have a definition:

An epithelial-mesenchymal transition (EMT) is a biologic process that allows a polarized epithelial cell, which normally interacts with basement membrane via its basal surface, to undergo multiple biochemical changes that enable it to assume a mesenchymal cell phenotype, which includes enhanced migratory capacity, invasiveness, elevated resistance to apoptosis, and greatly increased production of ECM (extra cellular matrix) components.

The completion of an EMT is signaled by the degradation of underlying basement membrane and the formation of a mesenchymal cell that can migrate away from the epithelial layer in which it originated.

Thus many cells are organized in a certain manner to effect certain functions. In the prostate, a glandular organ, there are basal and luminal cells surrounding the glands wherein secretion occurs. In the case of high grade PIN for example, the cells start to proliferate and no longer align properly. Then they slowly depart and create for wont of a better term, move out. They continue:

A number of distinct molecular processes are engaged in order to initiate an EMT and enable it to reach completion. These include activation of transcription factors, expression of specific cell-surface proteins, reorganization and expression of cytoskeletal proteins, production of ECM-degrading enzymes, and changes in the expression of specific microRNAs. In many cases, the involved factors are also used as biomarkers to demonstrate the passage of a cell through an EMT..

The pioneering work of Elizabeth Hay first described an “epithelial mesenchymal transformation” using a model of chick primitive streak formation. In the intervening time, the term “transformation” has been replaced with “transition,” reflecting in part the reversibility of the process and the fact that it is distinct from neoplastic transformation.

The phenotypic plasticity afforded by an EMT is revealed by the occurrence of the reverse process — a mesenchymal- epithelial transition (MET), which involves the conversion of mesenchymal cells to epithelial derivatives. Relatively little is known about this process; the best-studied example is the MET associated with kidney formation, which is driven by genes such as paired box 2 (Pax2), bone morphogenetic protein 7 (Bmp7), and Wilms tumor 1 (Wt1).

From Kalluri and Weinberg we have three types of MET cells are discussed:

(A) Type 1 EMT is associated with implantation and embryonic gastrulation and gives rise to the mesoderm and endoderm and to mobile neural crest cells. The primitive epithelium, specifically the epiblast, gives rise to primary mesenchyme via an EMT. This primary mesenchyme can be re-induced to form secondary epithelia by a MET. It is speculated that such secondary epithelia may further differentiate to form other types of epithelial tissues and undergo subsequent EMT to generate the cells of connective tissue, including astrocytes, adipocytes, chondrocytes, osteoblasts, and muscle cells.

(B) EMTs are re-engaged in the context of inflammation and fibrosis and represent the type 2 EMTs. Unlike the type 1 EMT, the type 2 EMT is expressed over extended periods of time and can eventually destroy an affected organ if the primary inflammatory insult is not removed or attenuated.

(C) Finally, the secondary epithelia associated with many organs can transform into cancer cells that later undergo the EMTs that enable invasion and metastasis, thereby representing type 3 EMTs.

Namely this details the three types; (i) those involved in a developing organism, (ii) those involved in a repairing organism, and (iii) those involved in a metastasizing organism. There is a similarity amongst these three.

As Kong et al have noted:

Cancer stem cells (CSCs) are cells within a tumor that possess the capacity to self-renew and maintain tumor-initiating capacity through differentiation into the heterogeneous lineages of cancer cells that comprise the whole tumor. These tumor-initiating cells could provide a resource for cells that cause tumor recurrence after therapy. Although the cell origin of CSCs remains to be fully elucidated, mounting evidence has demonstrated that Epithelial-to-Mesenchymal Transition (EMT), induced by different factors, is associated with tumor aggressiveness and metastasis and these cells share molecular characteristics with CSCs, and thus are often called cancer stem-like cells or tumor-initiating cells.

The acquisition of an EMT phenotype is a critical process for switching early stage carcinomas into invasive malignancies, which is often associated with the loss of epithelial differentiation and gain of mesenchymal phenotype. Recent studies have demonstrated that EMT plays a critical role not only in tumor metastasis but also in tumor recurrence and that it is tightly linked with the biology of cancer stem-like cells or cancer-initiating cells. Here we will succinctly summarize the state-of-our-knowledge regarding the molecular similarities between cancer stem-like cells or CSCs and EMT-phenotypic cells that are associated with tumor aggressiveness focusing on solid tumors.


We now consider several observations resulting for the above analysis. We examine four areas:

1. We look at the issue of cancer stem cells and their relationship to the EMT process. CSC are interesting targets of interest since targeting them may be much more effective than targeting bulk tumors. All too often removing a bulk tumor without regard to a CSC presence just means recurrence. It is often the case where a surgeon gets a clear margin on an excision and declares victory while a CSC has escaped.

2. Circulating tumor cells or parts thereof have become of significant interest in what has been termed liquid biopsies. Namely constituents of tumor cells in the blood can be detected and analyzed. Here we look at markers for excess EMT process.

3. There has been an evolving understanding of the EMT process. We briefly discuss this change.

4. The arear of new therapeutics is key. One specific area we have tried to open is based upon the following logic.

a)     EMT is related to and a putative driver of metastatic growth.
b)     EMT as a process is heavily influenced by immune system drivers
c)     Perhaps immunotherapeutic approaches to mitigating EMT processes may be effected and this down regulate any metastatic results.

Thus studying the EMT process can add significantly to our understanding of a multiplicity of cancers.

1. Cancer Stem Cells

Stem cells have been discussed at length in the context of many cancers. They are often closely associated with the EMT process. As Mitra et al note:

Tumor relapse and metastasis are the primary causes of poor survival rates in patients with advanced cancer despite successful resection or chemotherapeutic treatment. A primary cause of relapse and metastasis is the persistence of cancer stem cells (CSCs), which are highly resistant to chemotherapy. Although highly efficacious drugs suppressing several subpopulations of CSCs in various tissue-specific cancers are available, recurrence is still common in patients. To find more suitable therapy for relapse, the mechanisms underlying metastasis and drug-resistance associated with relapse-initiating CSCs need to be identified. Recent studies in circulating tumor cells (CTCs) of some cancer patients manifest phenotypes of both CSCs and epithelial-mesenchymal transition (EMT).

These patients are unresponsive to standard chemotherapies and have low progression free survival, suggesting that EMT-positive CTCs are related to co-occur with or transform into relapse-initiating CSCs.

Furthermore, EMT programming in cancer cells enables in the remodeling of extracellular matrix to break the dormancy of relapse-initiating CSCs. In this review, we extensively discuss the association of the EMT program with CTCs and CSCs to characterize a subpopulation of patients prone to relapses.

Identifying the mechanisms by which EMT-transformed CTCs and CSCs initiate relapse could facilitate the development of new or enhanced personalized therapeutic regimens.

We have discussed the CSC construct especially in the case of PCa. It could be argued that identifying the PCa and removing them would then make any of the other cells indolent.  CSC development still is a complex area. Just how a CSC is formed and how it manages to survive and prosper is complex. Perhaps the nexus with the EMT process may assist in better understanding.


Circulating Tumor Cells, CTC, and parts therefrom, such as RNA fragments, even DNA fragments, are also a current topic of interest in detecting and monitoring cancers. Since EMT is considered an essential part of the metastatic process, then it would seem logical to also look for EMT markers as well.

As Heerboth et al note:

Another exciting area of research is the use of EMT markers in the analysis of circulating tumor cells (CTC). Diagnostically, CTC has been a mainstay of clinical practice in assessment of metastasis and prognosis. The presence of CTC in a patient’s blood can be measured using the AdnaTest, a PCR assay for markers of EMT such as Twist, Akt, and Pi3k. The test employs a method for enriching the CTCs in a blood sample using antibodies conjugated to magnetic beads. Once the tumor cells have been pulled down, the mRNA can be isolated and expression of EMT markers determined. The test is reported to be sensitive enough to detect two CTCs in a 5 mL sample of blood.

Recent works have indicated that consideration of CTC EMT status is critical to achieve a more accurate prognosis. In studies of metastatic breast cancer, CTC were found to express known EMT regulators, including TGF-β pathway components and the FOXC1 transcription factor. These data support a role for EMT in the blood-borne dissemination of human breast cancer. Classical markers of EMT, Twist, and vimentin, have been identified in breast cancer patients and specifically show elevated expression in patients with metastatic cancer relative to patients with early stage cancer, supporting the hypothesis that EMT controls the metastatic potential of CTCs

Thus we see that a more complex set of blood borne markers may be identified and profiled to establish cancer diagnosis, prognosis and arguably even fine tuning on therapeutics and therapeutic targeting.

From Lee et al we have a list of putative markers. Whether any of these are specifically appropriate will take time to study. The issue one assumes is to better understand EMT as it pertains to a malignancy. For example, for decades in breast cancer, in melanoma, and other cancers, removal of lymph nodes was considered standard practice even if no overt sign of metastasis was present. The resulting morbidity was often significant. If however one seeks EMT processes then perhaps one may attain a more viable and specific alternative.


There has been a debate over the years regarding the nature of EMT and cancer. As Tian (2005) notes in an earlier paper:

Epithelial mesenchymal transition has been postulated as a versatile mechanism which facilitates cellular repositioning and redeployment during embryonic development, tissue reconstruction after injury, carcinogenesis, and tumor metastasis. The hypothesis originates from parallels drawn between the morphology and behavior of locomotory and sedentary cells in vitro and in various normal and pathologic processes in vivo.

This review analyzes data from several studies on embryonic development, wound healing, and the pathology of human tumors, including work from our own laboratory, to assess the validity of the proposal. It is concluded that there is no convincing evidence for conversion of epithelial cells into mesenchymal cell lineages in vivo and that the biological repertoire of normal and malignant cells is sufficient to account for the events and processes observed, without needing to invoke radical changes in cell identity.

The author then goes on with a detailed "on the other hand" discussion of EMT relevance. This is always a worthwhile analysis to come back to from time to time.

However Roche (2018) notes some thirteen years later:

The epithelial-to-mesenchymal transition (EMT) occurs during normal embryonic development, tissue regeneration, organ fibrosis, and wound healing. It is a highly dynamic process, by which epithelial cells can convert into a mesenchymal phenotype. However, it is also involved in tumor progression with metastatic expansion, and the generation of tumor cells with stem cell properties that play a major role in resistance to cancer treatment.

EMT is not complete in cancer cells, and tumor cells are in multiple transitional states and express mixed epithelial and mesenchymal genes.

Such hybrid cells in partial EMT can move collectively as clusters, and can be more aggressive than cells with a complete EMT phenotype. EMT is also reversible by the mesenchymal-to-epithelial transition (MET), thought to affect circulating cancer cells when they reach a desirable metastatic niche to develop secondary tumors. The EMT process involves the disruption of cell–cell adhesion and cellular polarity, remodeling of the cytoskeleton, and changes in cell–matrix adhesion. It is associated with improvement in migratory and invasive properties.

In cancers, EMT inducers are hypoxia, cytokines, and growth factors secreted by the tumor microenvironment, stroma crosstalk, metabolic changes, innate and adaptive immune responses, and treatment with antitumor drugs. Switch in gene expression from epithelial to mesenchymal phenotype is triggered by complex regulatory networks involving transcriptional control with SNAI1 and SNAI2, ZEB1 and ZEB2, Twist, and E12/E47 among transcriptional factors, non-coding RNAs (miRNAs and long non-coding RNAs), chromatin remodeling and epigenetic modifications, alternative splicing, post-translational regulation, protein stability, and subcellular localization.

EMT is becoming a target of interest for anticancer therapy. However, more knowledge about the role of EMT in metastasis, its control, and its reversion is necessary. Indeed, alternative modes of dissemination, colonization via a MET-independent pathway, and investigation of circulating cancer cells in the blood support a more nuanced view of the role of EMT and MET in cancer metastasis.

The above argument seems to strengthen the assertion of the significance of EMT and as importantly the MET reversal process which we have discussed.


The understanding of the EMT process presents opportunities for therapeutic development. Mladinich et al have noted:

Cancer stem cell (CSC) has become recognized for its role in both tumorigenesis and poor patient prognosis in recent years. Traditional therapeutics are unable to effectively eliminate this group of cells from the bulk population of cancer cells, allowing CSCs to persist posttreatment and thus propagate into secondary tumors. The therapeutic potential of eliminating CSCs, to decrease tumor relapse, has created a demand for identifying mechanisms that directly target and eliminate cancer stem cells. Molecular profiling has shown that cancer cells and tumors that exhibit the CSC phenotype also express genes associated with the epithelial-to-mesenchymal transition (EMT) feature.

Ample evidence has demonstrated that upregulation of master transcription factors (TFs) accounting for the EMT process such as Snail/Slug and Twist can reprogram cancer cells from differentiated to stem-like status. Despite being appealing therapeutic targets for tackling CSCs, pharmacological approaches that directly target EMT-TFs remain impossible. In this review, we will summarize recent advances in the regulation of Snail/Slug and Twist at transcriptional, translational, and posttranslational levels and discuss the clinical implication and application for EMT blockade as a promising strategy for CSC targeting.

Thus there may be avenues of access to controlling the CSC via the EMT process. The authors conclude:

These studies indicate that approaches which inhibit protein expression or activity upstream of EMT-TFs will have a better chance to achieve CSC eradiation. Extensive work as reviewed above shed light on new approaches for the targeting of EMT-TFs. As our understanding of protein regulation of EMT-TFs advances, the ability to generate or repurpose new candidate molecules to target CSCs increases.

Specific inactivation of EMT-TFs in combination with chemotherapy will likely enhance patient survival long-term via targeting of both CSCs and differentiated tumor cells. We have reasons for optimism that future studies on structural information of upstream regulators of EMT-TFs and on the crosstalk between upstream regulators and EMT-TFs would yield new CSC therapeutics.



1.     Biechele, T., et al, Wnt/β-Catenin Signaling and AXIN1 Regulate Apoptosis Triggered by Inhibition of the Mutant Kinase BRAFV600E in Human Melanoma, Sci. Signal., 10 January 2012 Vol. 5, Issue 206, p. ra3.
2.     Cai, C., et al, ERG induces androgen receptor-mediated regulation of SOX9 in prostate cancer, The Journal of Clinical Investigation http://www.jci.org  Volume 123 Number 3 March 2013.
3.     Chen, Y., et al, TMPRSS2, a Serine Protease Expressed in the Prostate on the Apical Surface of Luminal Epithelial Cells and Released into Semen in Prostasomes, Is Misregulated in Prostate Cancer Cells, The American Journal of Pathology, Vol. 176, No. 6, June 2010
4.     Demichelis, F., et al, Distinct Genomic Aberrations Associated with ERG Rearranged Prostate Cancer, Gene, Chromo, Cancer, V 48, 1999, pp 366-380.
5.     Demichelis, F., et al, TMPRSS2:ERG Gene Fusion Associated with Lethal Prostate Cancer in a Watchful Waiting Cohort, Onco 2007 pp. 1-4.
6.     DeNunzio, et al, The Controversial Relationship Between Benign Prostatic Hyperplasia and Prostate Cancer: The Role of Inflammation, Euro Uro 2011.
7.     Dobi, A., et al, ERG Expression Levels in Prostate Tumors Reflect Functional Status of the Androgen Receptor, Open Cancer Jrl, V 3, 2010, pp 101-108.
8.     Esgueva, R., et al, Prevalanece of TMPRSS2-ERG and SLC45A3-ERG Gene Fusions in a Large Prostatectomy Cohort, Mod Path, V  2010, pp 1-8.
9.     Fenouille, et al, The Epithelial-Mesenchymal Transition (EMT) Regulatory Factor SLUG (SNAI2) Is a Downstream Target of SPARC and AKT in Promoting Melanoma Cell Invasion, PLoS ONE, www.plosone.org , 1 July 2012, Volume 7, Issue 7, e40378
10.  Flajollet , S. et al, Abnormal Expression of the ERG Transcription Factor in Prostate Cancer Cells Activates Osteopontin, Mol Cancer Res; 9(7) July 2011.
11.  Goldstein, A. et al, Identification of a Cell of Origen for Human Prostate Cancer, Science, 2010 V 329, pp 568-571.
12.  Gong et al, Epigenetic silencing of TET2 and TET3 induces an EMT-like process in melanoma, Oncotarget, 2017, Vol. 8, (No. 1), pp: 315-328
13.  Goss, K., M. Kahn, Targeting the Wnt Pathway in Cancer, Springer (New York) 2011.
14.  Hearing V., S. Leong, From Melanocytes to Melanoma, Humana 2011.
15.  Heerboth et al, EMT and tumor metastasis, Clinical and Translational Medicine (2015) 4:6
16.  Iljin, K., et al, TMPRSS2 Fusions with Oncogenes ETS Factors in Prostate Cancer, Cancer Res, V 66, 2006, pp 10242-10246.
17.  Jagle et al, SNAIL1-mediated downregulation of FOXA proteins facilitates the inactivation of transcriptional enhancer elements at key epithelial genes in colorectal cancer cells, PLOS Genetics November 20, 2017
18.  Jin, H., et al, Androgen Receptor-Independent Function of FoxA1 in Prostate Cancer Metastasis, Cancer Res; 73(12) June 15, 2013
19.  Jue et al, Notch1 promotes vasculogenic mimicry in hepatocellular carcinoma by inducing EMT signaling, Oncotarget, 2017, Vol. 8, (No. 2), pp: 2501-2513
20.  Kalluri and Weinberg, The basics of epithelial-mesenchymal transition, The Journal of Clinical Investigation http://www.jci.org Volume 119 Number 6 June 2009
21.  Kalluri, EMT: When epithelial cells decide to become mesenchymal-like cells, The Journal of Clinical Investigation http://www.jci.org Volume 119 Number 6 June 2009
22.  King, J., Cooperativity of TMPRSS2-ERG with PI3 kinase pathway Activation in Prostate Oncogenesis, Nature Gen, V 41, 2009, pp 524-526.
23.  Kong et al, Cancer Stem Cells and Epithelial-to-Mesenchymal Transition (EMT)-Phenotypic Cells: Are They Cousins or Twins? Cancers 2011, 3, 716-729
24.  Kong et al, Epithelial to Mesenchymal Transition Is Mechanistically Linked with Stem Cell Signatures in Prostate Cancer Cells, PLOS ONE, August 2010, Volume 5, Issue 8, e12445
25.  Lee et al, The epithelial–mesenchymal transition: new insights in signaling, development, and disease, The Journal of Cell Biology, Vol. 172, No. 7, March 27, 2006 973–981
26.  Lippolis, G., et al, A high-density tissue microarray from patients with clinically localized prostate cancer reveals ERG and TATI exclusivity in tumor cells, Prostate Cancer and Prostatic Disease (2013) 16, 145–150.
27.  Mitra et al, EMT, CTCs and CSCs in tumor relapse and drug-resistance, Oncotarget, Vol. 6, No. 13, 2015
28.  Mladinich et al, Tackling Cancer Stem Cells via Inhibition of EMT Transcription Factors, Stem Cells International, Volume 2016, Article ID 5285892
29.  Mosquera, J., et al, Characterization of TMPRSS2-ERG Fusion in High Grade Prostatic Intraepithelial Neoplasia and Potential Clinical Implications, Clin Can Res, V 14, 2008, pp 3380-3385.
30.  Mosquera, J., et al, Morphological Features of TMPRSS2-ERG Gene Fusion Prostate Cancer, Jrl Path 2007 pp 91-101.
31.  Mosquera, J., et al, Prevalence of TMPRSS2-ERG Fusion Prostate Cancer among Men Undergoing Prostate Biopsy in the United States, Clin Can Res, V 15, 2009, 4706-4711.
32.  Mulholland et al, Pten Loss and RAS/MAPK Activation Cooperate to Promote EMT and Metastasis Initiated from Prostate Cancer Stem Progenitor Cells, Cancer Res; 72(7) April 1, 2012
33.  Murphy, M., Diagnostic and Prognostic Biomarkers and Theraputic Targets in Melanoma, Humana (Springer, New York), 2012.
34.  Park, K., et al, TMPRSS2:ERG Gene Fusion Predicts Subsequent Detection of Prostate Cancer in Patients With High-Grade Prostatic Intraepithelial Neoplasia, JCO December 2, 2013.
35.  Protopsaltis, I., et al, Linking Pre-Diabetes with Benign Prostate Hyperplasia. IGFBP-3: A Conductor of Benign Prostate Hyperplasia Development Orchestra, PLOS ONE, www.plosone.org, 1 December 2013, Volume 8, Issue 12. http://www.plosone.org/article/fetchObject.action?uri=info%3Adoi%2F10.1371%2Fjournal.pone.0081411&representation=PDF
36.  Radisky, Epithelial-mesenchymal transition, Journal of Cell Science 118 (19)
37.  Reddy S., et al, The erg gene: A human gene related to the ets oncogene, Proc. Nati. Acad. Sci. USA, Vol. 84, pp. 6131-6135, September 1987.
38.  Roche, The Epithelial-to-Mesenchymal Transition in Cancer, Cancers 2018, 10, 52
39.  Rubin, M., A. Chinnaiyan, Bioinformatics approach leads to the discovery of the TMPRSS2:ETS gene fusion in prostate cancer, Laboratory Investigation (2006) 86, 1099–1102.
40.  Rubin, M., A. Chinnaiyan, Bioinformatics Approach Leads to the Discovery of the TMPRSS2:ETS gene Fusion in Prostate Cancer, Lab Inv 2006, pp. 1099-1102.
41.  Rubin, M., et al, Overexpression Amplification and Androgen Regulation of TPD52 in Prostate Cancer, Can Res 2004 pp 3814-3822.
42.  Savagner, The epithelial–mesenchymal transition (EMT) phenomenon, Annals of Oncology 21 (Supplement 7): vii89–vii92, 2010
43.  Shih and Yang, The EMT regulator slug and lung carcinogenesis, Carcinogenesis vol.32 no.9 pp.1299–1304, 2011
44.  Shtivelman et al, Molecular pathways and targets in prostate cancer, Oncotarget, Vol. 5, No. 17, 2014
45.  Song et al, Loss of FOXA1/2 Is Essential for the Epithelial-to-Mesenchymal Transition in Pancreatic Cancer, AACT Jrl 2010
46.  Suarez-Carmona et al, EMT and inflammation: inseparable actors of cancer progression, Molecular Oncology 11 (2017) 805–823, 2017
47.  Takahashi et al, The role of microRNAs in the regulation of cancer stem cells, Frontiers in Genetics, January2014|Volume4|Article295 | 1
48.  Thompson and Newgreen, Carcinoma Invasion and Metastasis: A Role for Epithelial-Mesenchymal Transition?, Cancer Res 2005; 65: (14). July 15, 2005
49.  Tian, The Fallacy of Epithelial Mesenchymal Transition in Neoplasia, Cancer Res 2005; 65: (14). July 15, 2005
50.  Tomlins, A., ETS Rearrangements and Prostate Cancer Initiation, Nature, V 448, 2007, pp 595-599.
51.  Tomlins, S., et al, ETS Gene Fusion in Prostate Cancer, Eur Jrl Uro 2009 pp 1-12.
52.  Tomlins, S., et al, Recurrent Fusion of TMPRSS2 and ETS Transcription Factor Genes in Prostate Cancer, Science, V 310, 2005, pp 644-648.
53.  Tomlins, S., et al, Role of the TMPRSS2–ERG Gene Fusion in Prostate Cancer, Neoplasia, Volume 10 Number 2 February 2008 pp. 177–188.
54.  Tsuji et al, Epithelial-Mesenchymal Transition and Cell Cooperativity in Metastasis, Cancer Res 2009; 69: (18). September 15, 2009
55.  Vikash et al, Glomerular parietal epithelial cells of adult murine kidney undergo EMT to generate cells with traits of renal progenitors, J. Cell. Mol. Med. Vol 15, No 2, 2011 pp. 396-413
56.  Wang et al, Role of tumor microenvironment in tumorigenesis, Journal of Cancer, 2017; 8(5): 761-773
57.  Wehbe et al, Epithelial-Mesenchymal-Transition-Like and TGFb Pathways Associated with Autochthonous Inflammatory Melanoma Development in Mice, PLOS One Nov 2012 Vol 7 Issue 11
58.  Weinberg, Cancer, 2nd Ed, Garland (New York) 2014
59.  Weinberg, R., The Biology of Cancer, Garland (New York) 2008.
60.  Wu, L., et al, ERG Is a Critical Regulator of Wnt/LEF1 Signaling in Prostate Cancer, Cancer Res October 1, 2013 73; 6068. http://cancerres.aacrjournals.org/content/73/19/6068.figures-only
61.  Wyatt, A., M. Gleave, Targeting the adaptive molecular landscape of castration-resistant prostate cancer, EMBO Molecular Medicine, April 2015.
62.  Xie et al, Role of DAB2IP in modulating epithelial-to-mesenchymal transition and prostate cancer metastasis, PNAS | February 9, 2010 | vol. 107 | no. 6 | 2485–2490
63.  Yang et al, Twist, a Master Regulator of Morphogenesis, Plays an Essential Role in Tumor Metastasis, Cell, Volume 117, Issue 7, 25 June 2004, Pages 927-939
64.  Yuan, L. et al, ETS-related Gene (ERG) Controls Endothelial Cell Permeability via Transcriptional Regulation of the Claudin 5 (CLDN5) Gene, J. Biol. Chem. 2012, 287:6582-6591.
65.  Zeisberg and Neilson, Biomarkers for epithelial-mesenchymal transitions, The Journal of Clinical Investigation http://www.jci.org  Volume 119 Number 6 June 2009 1429
66.  Zhao et al, Inhibition of Snail Family Transcriptional Repressor 2 (SNAI2) Enhances Multidrug Resistance of Hepatocellular Carcinoma Cells, PLOS, October 19, 2016
67.  Zong, Y., et al, ETS Family Transcription Factors Collaborate with Alternative Signalling Pathways to Induce Carcinomas from Adult Murine Prostate Cells, PNAS, V 106, 209, pp 12465-12470.




[1] Weinberg pp 657-669