958 resultados para Androgen Independance, Castration-Resistant, Androgen Receptor, shRNA, Tumor Progression
Resumo:
Neuroendocrine differentiation has been described in rectal adenocarcinomas receiving neoadjuvant therapy prior to radical surgery, but its clinical relevance is controversial and no data are currently available in colorectal carcinoma metastases as compared to primary tumors. The presence of chromogranin A positive tumor cells was investigated by means of immunohistochemistry on surgical specimens from 54 primary colorectal carcinomas and their corresponding metastases, resected at diagnosis or during tumor progression. In 47 patients, tumor metastases were resected 1 month to 12 years after chemotherapy and/or radiotherapy, while in the remaining seven patients no additional therapy after primary surgery was performed. In primary tumors, neuroendocrine differentiation was found in 12/54 cases (22.2%) as compared to 25/54 metastatic lesions (46.3%; p?=?0.01). The presence of neuroendocrine phenotype was not correlated with any clinical pathological parameter nor with a different proliferation index. However, patients having neuroendocrine cells in the primary tumor had a significantly shorter survival from the time of metastatic spread than those having not (33.3 vs. 55.5 months; p?=?0.04). In summary, our data show that colorectal carcinoma metastases contain a higher percentage of neuroendocrine differentiated cells as compared to their corresponding primaries, a finding possibly related to the influence of chemotherapy in neuroendocrine differentiation during colorectal carcinoma progression.
Resumo:
Abstract The c-myc gene is one of the most frequently mutated oncogenes found in human tumors. c-Myc has been implicated in the regulation of various biological processes including cell cycle progression, cellular growth, differentiation, angiogenesis, immortalization and apoptosis. To assess the normal role of c-Myc in epithelial cell types in vitro and in vivo we have deleted the c-myc gene in keratinocytes and in the adult skin epidermis by conditional Cre/loxP mediated recombination. Similar to what we have previously shown in mouse embryonic fibroblasts acute elimination of c-Myc activity in cultured keratinocytes causes cells to cease proliferation and adapt a flat cell morphology. Mutant cells accumulate in a diploid Ki67neg stage, indicative of a quiescent Go stage. This demonstrates that c-Myc activity is essential to maintain keratinocytes in a productive cell cycle. In addition, mutant keratinocytes showed a defect in Ca2+ induced induction of the differentiation marker Keratin 1 suggesting a role for c-Myc during differentiation. To assess the in vivo role of c-Myc we used a tamoxifen inducible K5::CreERT transgene to delete the c-myc gene in the adult skin epidermis. Unexpectedly, despite strong c-Myc expression in the basal compartment it is not required for maintenance of the skin epidermis in the adult mouse. The epidermis appeared normal with respect to both proliferation and differentiation. In addition, no selection against c-Myc deficient epidermal cells occurred over many months, further confirming that c-Myc is dispensable for normal skin homeostasis. Even more surprising, TPA induced hyperproliferation also occurred in a c-Myc independent manner. Treatment of the skin with the mutagen DMBA prior to TPA is a classical way to induce papillomas by selecting for mutations that lead to dominant activation of the oncogene Ha-Ras. Most interestingly tumor formation was severely inhibited suggesting that tumor progression requires endogenous c-Myc. Further studies are required to address whether the role of c-Myc in the activation of telomerase or the Werner protein, or its role to induce angiogenesis is required for skin tumor progression, In conclusion, this work shows that while c-Myc is not required for maintenance or hyperplasia of mouse epidermis, it is essential for skin tumor progression in collaboration with Ras. Résumé Le gène c-myc est un des oncogènes les plus fréquemment mutés dans les tumeurs humaines. c-Myc est impliqué dans la régulation de processus biologiques variés, comme la progression du cycle cellulaire, la croissance cellulaire, la différenciation, l'angiogenèse, l'immortalisation et l'apoptose. Pour caractériser le rôle physiologique de c-Myc dans les cellules de type épithélial in vitro et in vivo, le gène c-myc a été délété dans des kératinocytes primaires et dans l'épiderme de peau de souris adultes par des recombinaisons conditionnelles (système Cre/loxP). De la même façon que dans les fibroblastes d'embryon de souris, l'élimination aiguë de l'activité de c-Myc dans les kératinocytes en culture primaire provoque l'arrêt de la prolifération des cellules et leur applatissement morphologique. Les cellules mutantes restent dans un stade diploïde Ki67neg, indiquant un stade quiescent Go. Cela démontre que l'activité de c-Myc est essentielle pour maintenir les kératinocytes dans le cycle cellulaire. De plus, les kératinocytes mutants montrent une déficience pour le marqueur de différenciation Kératine 1 au cours de la différenciation induite par le calcium, suggérant un rôle de c-Myc dans la différenciation cellulaire. Pour comprendre le rôle de c-Myc in vivo, le transgène K5::CreERT inductible par le tamoxifen a été utilisé pour déléter le gène c-inyc dans l'épiderme de souris adultes. Etonnemment, malgré une forte expression de c-Myc dans le compartiment basal de l'épiderme, ce gène n'est pas nécessaire pour la maintenance de l'épiderme de la peau chez la souris adulte. L'épiderme apparait normal avec une prolifération et une différenciation physiologique des cellules. De plus, il n'y a pas de sélection contre les cellules épidennales c-Myc déficientes après plusieurs mois, ce qui confirme que c-Myc n'est pas nécessaire pour l'homéostasie normale de la peau. Encore plus surprenant, une hyperprolifération est également induite par du TPA chez les souris mutantes, impliquant une voie de prolifération indépendante de c-Myc. Le traitement de la peau par le mutagène DMBA avant le traitement au TPA est une voie classique d'induction de papillomes, par sélection de mutations conduisant à l'activation de l'oncogène Ha-Ras. La formation des tumeurs est fortement inhibée chez les souris mutantes, suggérant que la progression des tumeurs nécessite la présence endogène de c-Myc. De nouvelles études sont nécessaires pour savoir si c-Myc a un rôle dans l'activation de la télomérase ou de la protéine de Werner, ou encore dans l'angiogénèse, qui sont nécessaires pour la progression tumorale. En conclusion, ce travail montre que même si c-Myc n'est pas nécessaire pour la maintenance ou l'hyperplasie de la peau de souris, il est essentiel pour la progression des tumeurs de la peau en collaboration avec Ras.
Resumo:
BACKGROUND New biomarkers are needed for the prognosis of advanced colorectal cancer, which remains incurable by conventional treatments. O6-methylguanine DNA methyltransferase (MGMT) methylation and protein expression have been related to colorectal cancer treatment failure and tumor progression. Moreover, the presence in these tumors of cancer stem cells, which are characterized by CD133 expression, has been associated with chemoresistance, radioresistance, metastasis, and local recurrence. The objective of this study was to determine the prognostic value of CD133 and MGMT and their possible interaction in colorectal cancer patients. METHODS MGMT and CD133 expression was analyzed by immunohistochemistry in 123 paraffin-embedded colorectal adenocarcinoma samples, obtaining the percentage staining and intensity. MGMT promoter methylation status was obtained by using bisulfite modification and methylation-specific PCR (MSP). These values were correlated with clinical data, including overall survival (OS), disease-free survival (DFS), tumor stage, and differentiation grade. RESULTS Low MGMT expression intensity was significantly correlated with shorter OS and was a prognostic factor independently of treatment and histopathological variables. High percentage of CD133 expression was significantly correlated with shorter DFS but was not an independent factor. Patients with low-intensity MGMT expression and ≥50% CD133 expression had the poorest DFS and OS outcomes. CONCLUSIONS Our results support the hypothesis that MGMT expression may be an OS biomarker as useful as tumor stage or differentiation grade and that CD133 expression may be a predictive biomarker of DFS. Thus, MGMT and CD133 may both be useful for determining the prognosis of colorectal cancer patients and to identify those requiring more aggressive adjuvant therapies. Future studies will be necessary to determine its clinical utility.
Resumo:
Summary : Clinical evidence indicates that tumors recurring within previously irradiated fields are highly invasive and metastatic, suggesting a role of the tumor stroma in this effect. Angiogenesis plays a critical role in tumor progression. Ionizing radiation is known to induce apoptosis of angiogenic endothelial cells, while the effect on quiescent endothelial cells and de novo angiogenesis is not well characterized. We recently observed that irradiation of normal tissue prevents tumor- and growth factor-induced angiogenesis. The main aim of my thesis work was to characterize the mechanisms of radiation-mediated inhibition of angiogenesis. To this purpose we used a combination of in vivo and ex vivo studies on irradiated healthy tissue, and in vitro irradiation experiments using angiogenesis models and isolated endothelial cells. We found that irradiation did not induce endothelial cell apoptosis and did not disrupt quiescent vessels within irradiated skin. Radiation reduced the recruitment of leukocytes to angiogenic Matrigel plugs, but this effect was rather secondary to decreased angiogenesis, as exogenous addition of leucocytes to Matrigel plugs did not rescue the angiogenesis defects. To ascertain the direct effect of radiation on endothelial cells, we used the mouse aortic ring assay to test the sprouting capacity of irradiated endothelial cells ex vivo and in vitro, and found that irradiation completely suppressed endothelial cell sprouting. Using HUVEC cells, we showed that irradiation of quiescent confluent endothelial cells did not induce cell death but suppressed subsequent migration and cell proliferation and induced senescence. By Western blotting, we observed a rapid and sustained increase in p21 levels, previously shown to be activated by p53 in response to double strand break, and mediating senescence in human cells. Current experiments focus on the mechanism of sustained p21 upregulation and its role in reduced migration. Inhibition of endothelial cell migration and proliferation by radiation may explain reduced angiogenesis in tumors growing in previously irradiated fields.
Resumo:
The application of angiogenesis inhibitors in neurooncology is increasing. Initially, these drugs seemed to be very promising because of the surprisingly high neuroradiological response rates that were observed in first clinical trials. Meanwhile, this enthusiasm is waning, as the high response rates did not translate into substantial improvements in progression-free and overall survival. Tumor progression during or after antiangiogenic therapy is often associated with rapid clinical neurological deterioration and sometimes even with diffuse infiltrative gliomatosis-like neuroradiological phenotypes. Thus, the characterization and understanding of escape mechanisms are needed. The identification of criteria for defining the personalized use of angiogenesis inhibitors remains a challenge.
Resumo:
PURPOSE: We have investigated the expression and regulation of 15-hydroxyprostaglandin dehydrogenase (15-PGDH) in gastric cancer. EXPERIMENTAL DESIGN: Clinical gastric adenocarcinoma samples were analyzed by immunohistochemistry and quantitative real-time PCR for protein and mRNA expression of 15-PGDH and for methylation status of 15-PGDH promoter. The effects of interleukin-1beta (IL-1beta) and epigenetic mechanisms on 15-PGDH regulation were assessed in gastric cancer cell lines. RESULTS: In a gastric cancer cell line with a very low 15-PGDH expression (TMK-1), the 15-PGDH promoter was methylated and treatment with a demethylating agent 5-aza-2'-deoxycytidine restored 15-PGDH expression. In a cell line with a relatively high basal level of 15-PGDH (MKN-28), IL-1beta repressed expression of 15-PGDH mRNA and protein. This effect of IL-1beta was at least in part attributed to inhibition of 15-PGDH promoter activity. SiRNA-mediated knockdown of 15-PGDH resulted in strong increase of prostaglandin E(2) production in MKN-28 cells and increased cell growth of these cells by 31% in anchorage-independent conditions. In clinical gastric adenocarcinoma specimens, 15-PGDH mRNA levels were 5-fold lower in gastric cancer samples when compared with paired nonneoplastic tissues (n = 26) and 15-PGDH protein was lost in 65% of gastric adenocarcinomas (n = 210). CONCLUSIONS: 15-PGDH is down-regulated in gastric cancer, which could potentially lead to accelerated tumor progression. Importantly, our data indicate that a proinflammatory cytokine linked to gastric carcinogenesis, IL-1beta, suppresses 15-PGDH expression at least partially by inhibiting promoter activity of the 15-PGDH gene.
Resumo:
The regulation of the immune system is controlled by many cell surface receptors. A prominent representative is the 'molecular switch' HVEM (herpes virus entry mediator) that can activate either proinflammatory or inhibitory signaling pathways. HVEM ligands belong to two distinct families: the TNF-related cytokines LIGHT and lymphotoxin-α, and the Ig-related membrane proteins BTLA and CD160. HVEM and its ligands have been involved in the pathogenesis of various autoimmune and inflammatory diseases, but recent reports indicate that this network may also be involved in tumor progression and resistance to immune response. Here we summarize the recent advances made regarding the knowledge on HVEM and its ligands in cancer cells, and their potential roles in tumor progression and escape to immune responses. Blockade or enhancement of these pathways may help improving cancer therapy.
Resumo:
Transforming growth factor-beta (TGF-beta) and its related proteins regulate broad aspects of body development, including cell proliferation, differentiation, apoptosis and gene expression, in various organisms. Deregulated TGF-beta function has been causally implicated in the generation of human fibrotic disorders and in tumor progression. Nevertheless, the molecular mechanisms of TGF-beta action remained essentially unknown until recently. Here, we discuss recent progress in our understanding of the mechanism of TGF-beta signal transduction with respect to the regulation of gene expression, the control of cell phenotype and the potential usage of TGF-beta for the treatment of human diseases.
Resumo:
PURPOSE: The aim of this study was to assess the outcome of patients with primary spinal myxopapillary ependymoma (MPE). MATERIALS AND METHODS: Data from a series of 85 (35 females, 50 males) patients with spinal MPE were collected in this retrospective multicenter study. Thirty-eight (45%) underwent surgery only and 47 (55%) received postoperative radiotherapy (RT). Median administered radiation dose was 50.4 Gy (range, 22.2-59.4). Median follow-up of the surviving patients was 60.0 months (range, 0.2-316.6). RESULTS: The 5-year progression-free survival (PFS) was 50.4% and 74.8% for surgery only and surgery with postoperative low- (<50.4 Gy) or high-dose (>or=50.4 Gy) RT, respectively. Treatment failure was observed in 24 (28%) patients. Fifteen patients presented treatment failure at the primary site only, whereas 2 and 1 patients presented with brain and distant spinal failure only. Three and 2 patients with local failure presented with concomitant spinal distant seeding and brain failure, respectively. One patient failed simultaneously in the brain and spine. Age greater than 36 years (p = 0.01), absence of neurologic symptoms at diagnosis (p = 0.01), tumor size >or=25 mm (p = 0.04), and postoperative high-dose RT (p = 0.05) were variables predictive of improved PFS on univariate analysis. In multivariate analysis, only postoperative high-dose RT was independent predictors of PFS (p = 0.04). CONCLUSIONS: The observed pattern of failure was mainly local, but one fifth of the patients presented with a concomitant spinal or brain component. Postoperative high-dose RT appears to significantly reduce the rate of tumor progression.
Resumo:
Lipid mediators can trigger physiological responses by activating nuclear hormone receptors, such as the peroxisome proliferator-activated receptors (PPARs). PPARs, in turn, control the expression of networks of genes encoding proteins involved in all aspects of lipid metabolism. In addition, PPARs are tumor growth modifiers, via the regulation of cancer cell apoptosis, proliferation, and differentiation, and through their action on the tumor cell environment, namely, angiogenesis, inflammation, and immune cell functions. Epidemiological studies have established that tumor progression may be exacerbated by chronic inflammation. Here, we describe the production of the lipids that act as activators of PPARs, and we review the roles of these receptors in inflammation and cancer. Finally, we consider emerging strategies for therapeutic intervention.
Resumo:
RESUME La peau est un organe complex composé de deux parties distinctes: l'épiderme et le derme, séparé par une membrane basale. Dans la couche basale de l'épiderme, les melanocytes synthétisent la mélanine dans des mélanosomes. Les mélanosomes sont ensuite transportés des mélanocytes vers les kératinocytes, protégeant ainsi la peau des dégâts dus aux radiations U.V. La E-cadhérine assure l'adhésion entre les mélanocytes et les kératinocytes. Au cours de la transformation du mélanocyte en cellule malignes, les mélanocytes perdent l'expression de la E-cadhérine et, simultanément, se mettent à exprimer la N-cadhérine, ce phénomène est nommé « cadherin switch ». La perte de l'expression de la E-cadhérine permet au mélanocytes d'échapper au contrôle des kératinocytes, tandis que l'expression de la N-cadhérine promeut l'invasion métastasique des cellules de mélanome. Préalablement, nous avons trouvé qu'une fraction de la N-cadhérine était localisée les microdomaines membranaires spécialisés, enrichi en cholestérol et en glycosphingolipides, appelés « lipid rafts ». Une des particularité des « lipid rafts » est qu'ils sont riches en molécules permettant la transmission de signaux d'activation. De plus, des travaux récents rapportent qu'un sous-type de « lipid rafts » appelé caveolae pourrai contribuer à la progression tumorale. S'appuyant sur le rôle prépondérant de la N-cadhérine dans la progression du mélanome ainsi que sur sa présence dans les « lipid rafts », nous avons émis l'hypothèse que l'association de la N-cadhérine avec les « lipid rafts » pourrai contribuer à la progression du mélanome. Le but de ce projet à été de caractériser l'association de la Ncadhérine avec les « lipid rafts » au cours de la progression du mélanome. Au moyen de lignées cellulaires humaines, dérivées de mélanomes à différents stades de progression, nous avons trouvé que (1) la N-cadhérine est partiellement associée aux «lipid rafts » dans six lignées dérivées de mélanome en phase avancée de progression et dans des tumeurs expérimentales, mais pas dans deux lignées dérivées de mélanome à un stade plus précoce ; (2) l'association de la N-cadhérine dans les « lipid rafts » ne dépent pas de son niveau d'expression ; (3) la E-cadhérine n'est pas présente dans les « lipid rafts »d'une lignée de cellule de mélanome ayant conservé l'expression de la E-cadhérine ; (4) la localisation de la N-cadhérine dans les « lipid rafts »n'est pas modulée par les facteurs de croissance bFGF, IGF-I, et HRG1-β1, ni par des voies de signalisation impliquant MEK, PKA, les kinases de la famille Src, et PI3K ; (5) l'association de la N-cadhérine avec les « lipid rafts » n'est pas requise pour la stabilisation des jonctions adhérentes et n'est pas perturbée par la destruction de ces dernières ; (6) la N-cadhérine dans les « lipid rafts » forme un complexe avec β-caténine, p 120ctn et α-caténine. En conclusion, cette étude originale montre pour la première fois que dans des cellules de mélanome agressifs, une fraction de la N-cadhérine est localisée dans les « lipid rafts » en association avec β-caténine, p 120ctn et α-caténine. Comme la présence de la N-cadhérine dans les « lipid rafts » ne contribue pas à la formation de jonction adhérentes, cette étude suggère une nouvelle fonction pour la N-cadhérine dans les « lipid rafts ». SUMMARY Human skin is a complex organ composed of two layers separated by a basement membrane: the epidermis and the dermis. In the basal layer of the epidermis, the melanin-producing cells of the skin, the melanocytes deliver melanin-containing melanosomes to keratinocytes, thereby protecting the epidermis and the dermis from the deleterious effects of ultraviolet light. Melanocytes physically interact with keratinocytes through E-cadherin-mediated adhesion. During malignant transformation into melanoma cells, melanocytes lose E-cadherin expression and concomitantly gain expression of N-cadherin, a phenomenon referred to as "cadherin switch". Loss of E-cadherin allows melanocytes to escape the regulatory effects of neighbouring keratinocytes, while gain of N-cadherin expression promotes migration, invasion and metastatic abilities of melanoma cells. In preliminary experiments, we found that a fraction of N-cadherin localized to specialized membrane microdomains enriched in cholesterol- and glycosphingolipid, called lipid rafts. One particular feature of lipid rafts is that they are rich in signalling molecules and they possibly modulate transmembrane signalling events. Moreover, recent reports suggested that a specialized type of rafts called caveolae might contribute to tumor progression. Based on the documented role of N-cadherin in melanoma progression and its presence in lipid rafts of melanoma cells, we raised the hypothesis that the association of N-cadherin with lipid rafts might be relevant to melanoma progression. The aim of this project was to characterize N-cadherin associated to lipid rafts during melanoma progression. Using human melanoma cell lines derived from melanoma at different stages of progression, we found that (1) N-cadherin is partly associated to lipid rafts in six cell lines derived from melanomas at late stages of progression and in experimental tumors, but not in two melanoma cell lines derived from early stages; (2) N-cadherin targeting to lipid rafts does not depend on its expression level; (3) E-cadherin is not localized in lipid rafts of a melanoma cell line that retained E-cadherin expression; (4) N-cadherin localization to lipid rafts is not modulated by the growth factors bFGF, IGF-I, and HRG1-β1, nor by MEK-, PKA-, Src family kinases-, and PI3K-mediated signalling events; (5) the association of N-cadherin with lipid rafts is not required for adherens junctions stability nor it is perturbed by adherens junctions disruption; (6) N-cadherin in lipid rafts is in complex with β-catenin, p 120ctm and α-catenin. In conclusion, this study provides original evidence that in aggressive melanoma cells a pool of N-cadherin is localized in lipid rafts in association with β-catenin, p 120 and α-catenin. The presence of N-cadherin in lipid rafts independently of its involvement in adherens junctions formation, suggests a possible new role for N-cadherin recruited to lipid rafts. Further studies investigating the biological meaning of this localization promise to uncover new properties of this molecule.
Resumo:
The regulation of the immune system is controlled by many cell surface receptors. A prominent representative is the 'molecular switch' HVEM (herpes virus entry mediator) that can activate either proinflammatory or inhibitory signaling pathways. HVEM ligands belong to two distinct families: the TNF-related cytokines LIGHT and lymphotoxin-α, and the Ig-related membrane proteins BTLA and CD160. HVEM and its ligands have been involved in the pathogenesis of various autoimmune and inflammatory diseases, but recent reports indicate that this network may also be involved in tumor progression and resistance to immune response. Here we summarize the recent advances made regarding the knowledge on HVEM and its ligands in cancer cells, and their potential roles in tumor progression and escape to immune responses. Blockade or enhancement of these pathways may help improving cancer therapy.
Resumo:
Non-steroidal anti-inflammatory drugs (NSAIDs) and specific inhibitors of cyclooxygenase (COX)-2, are therapeutic groups widely used for the treatment of pain, inflammation and fever. There is growing experimental and clinical evidence indicating NSAIDs and COX-2 inhibitors also have anti-cancer activity. Epidemiological studies have shown that regular use of Aspirin and other NSAIDs reduces the risk of developing cancer, in particular of the colon. Molecular pathology studies have revealed that COX-2 is expressed by cancer cells and cells of the tumor stroma during tumor progression and in response to chemotherapy or radiotherapy. Experimental studies have demonstrated that COX-2 over expression promotes tumorigenesis, and that NSAIDs and COX-2 inhibitors suppress tumorigenesis and tumor progression. Clinical trials have shown that NSAIDs and COX-2 inhibitors suppress colon polyp formation and malignant progression in patients with familial adenomatous polyposis (FAP) syndrome. Recent advances in the understanding of the cellular and molecular mechanisms of the anti-cancer effects of NSAIDs and COX-2 inhibitors have demonstrated that these drugs target both tumor cells and the tumor vasculature. The therapeutic benefits of COX-2 inhibitors in the treatment of human cancer in combination with chemotherapy or radiotherapy are currently being tested in clinical trials. In this article we will review recent advances in the understanding of the anti-tumor mechanisms of these drugs and discuss their potential application in clinical oncology.
Resumo:
PURPOSE: This study was performed to determine the impact of perfusion and diffusion magnetic resonance imaging (MRI) sequences on patients during treatment of newly diagnosed glioblastoma. Special emphasis has been given to these imaging technologies as tools to potentially anticipate disease progression, as progression-free survival is frequently used as a surrogate endpoint. METHODS AND MATERIALS: Forty-one patients from a phase II temolozomide clinical trial were included. During follow-up, images were integrated 21 to 28 days after radiochemotherapy and every 2 months thereafter. Assessment of scans included measurement of size of lesion on T1 contrast-enhanced, T2, diffusion, and perfusion images, as well as mass effect. Classical criteria on tumor size variation and clinical parameters were used to set disease progression date. RESULTS: A total of 311 MRI examinations were reviewed. At disease progression (32 patients), a multivariate Cox regression determined 2 significant survival parameters: T1 largest diameter (p < 0.02) and T2 size variation (p < 0.05), whereas perfusion and diffusion were not significant. CONCLUSION: Perfusion and diffusion techniques cannot be used to anticipate tumor progression. Decision making at disease progression is critical, and classical T1 and T2 imaging remain the gold standard. Specifically, a T1 contrast enhancement over 3 cm in largest diameter together with an increased T2 hypersignal is a marker of inferior prognosis.
Resumo:
The ability of tumor cells to leave a primary tumor, to disseminate through the body, and to ultimately seed new secondary tumors is universally agreed to be the basis for metastasis formation. An accurate description of the cellular and molecular mechanisms that underlie this multistep process would greatly facilitate the rational development of therapies that effectively allow metastatic disease to be controlled and treated. A number of disparate and sometimes conflicting hypotheses and models have been suggested to explain various aspects of the process, and no single concept explains the mechanism of metastasis in its entirety or encompasses all observations and experimental findings. The exciting progress made in metastasis research in recent years has refined existing ideas, as well as giving rise to new ones. In this review we survey some of the main theories that currently exist in the field, and show that significant convergence is emerging, allowing a synthesis of several models to give a more comprehensive overview of the process of metastasis. As a result we postulate a stromal progression model of metastasis. In this model, progressive modification of the tumor microenvironment is equally as important as genetic and epigenetic changes in tumor cells during primary tumor progression. Mutual regulatory interactions between stroma and tumor cells modify the stemness of the cells that drive tumor growth, in a manner that involves epithelial-mesenchymal and mesenchymal-epithelial-like transitions. Similar interactions need to be recapitulated at secondary sites for metastases to grow. Early disseminating tumor cells can progress at the secondary site in parallel to the primary tumor, both in terms of genetic changes, as well as progressive development of a metastatic stroma. Although this model brings together many ideas in the field, there remain nevertheless a number of major open questions, underscoring the need for further research to fully understand metastasis, and thereby identify new and effective ways of treating metastatic disease.