Appearance
This report is written by MaltSci based on the latest literature and research findings
What are the mechanisms of cancer metastasis?
Abstract
Cancer metastasis is a multifaceted biological process that significantly contributes to cancer progression and is responsible for approximately 90% of cancer-related deaths globally. Unlike the well-characterized mechanisms of primary tumor formation, the pathways and factors driving metastasis remain poorly understood. Metastasis involves a series of complex steps, including the invasion of cancer cells from the primary tumor, intravasation into the bloodstream, survival in circulation, extravasation into distant tissues, and colonization at secondary sites. Each of these steps is regulated by intricate molecular and cellular mechanisms, influenced by the tumor microenvironment and various signaling pathways. Understanding cancer metastasis is crucial for developing effective therapeutic strategies aimed at preventing or treating metastatic disease. Recent advancements have highlighted the roles of genetic and epigenetic factors, cellular interactions within the tumor microenvironment, and molecular pathways facilitating metastatic progression. Key processes such as epithelial-mesenchymal transition (EMT) enhance the migratory and invasive capabilities of cancer cells, while the tumor microenvironment supports or inhibits metastatic spread. This review synthesizes current knowledge, exploring mechanisms of invasion, intravasation, survival in circulation, extravasation, and colonization. It discusses the tumor microenvironment's role, focusing on stromal interactions, extracellular matrix dynamics, and immune cell involvement. Additionally, the review examines molecular pathways regulating metastasis and current therapeutic strategies. Future research directions, including novel approaches and the importance of early detection, are emphasized. By providing a comprehensive overview of the mechanisms underlying cancer metastasis, this review aims to identify critical areas for future research and therapeutic intervention.
Outline
This report will discuss the following questions.
- 1 Introduction
- 2 Mechanisms of Cancer Metastasis
- 2.1 Invasion of Cancer Cells
- 2.2 Intravasation into Blood Vessels
- 2.3 Survival in Circulation
- 2.4 Extravasation and Colonization
- 3 Role of the Tumor Microenvironment
- 3.1 Interactions with Stromal Cells
- 3.2 Extracellular Matrix Dynamics
- 3.3 Immune Cell Involvement
- 4 Molecular Pathways in Metastasis
- 4.1 Key Signaling Pathways
- 4.2 Genetic and Epigenetic Regulation
- 5 Therapeutic Implications
- 5.1 Current Treatment Strategies
- 5.2 Emerging Therapeutic Targets
- 6 Future Directions in Metastasis Research
- 6.1 Novel Approaches and Technologies
- 6.2 Importance of Early Detection
- 7 Summary
1 Introduction
Cancer metastasis is a multifaceted biological process that significantly contributes to cancer progression and is responsible for approximately 90% of cancer-related deaths globally [1][2]. Unlike the well-characterized mechanisms of primary tumor formation, the pathways and factors driving metastasis remain poorly understood. Metastasis involves a series of complex steps, including the invasion of cancer cells from the primary tumor, intravasation into the bloodstream, survival in circulation, extravasation into distant tissues, and colonization at secondary sites [3][4]. Each of these steps is regulated by intricate molecular and cellular mechanisms, which are influenced by the tumor microenvironment and various signaling pathways [5][6].
The significance of understanding cancer metastasis cannot be overstated. The ability of cancer cells to disseminate and establish secondary tumors is a critical determinant of patient prognosis and treatment outcomes. Consequently, elucidating the mechanisms of metastasis is essential for the development of effective therapeutic strategies aimed at preventing or treating metastatic disease [7][8]. Recent advancements in cancer biology have shed light on the roles of genetic and epigenetic factors, cellular interactions within the tumor microenvironment, and the molecular pathways that facilitate metastatic progression [2][9].
Current research has identified key processes such as epithelial-mesenchymal transition (EMT), which endows cancer cells with enhanced migratory and invasive capabilities [5][10]. Furthermore, the tumor microenvironment, comprising various stromal cells, extracellular matrix components, and immune cells, plays a pivotal role in supporting or inhibiting metastatic spread [1][4]. Understanding these interactions and the molecular underpinnings of metastasis is crucial for identifying potential therapeutic targets and improving clinical outcomes for patients with metastatic cancer [5][9].
This review will be organized into several key sections. We will first explore the mechanisms of cancer metastasis, detailing the processes of invasion, intravasation, survival in circulation, extravasation, and colonization [3][5]. Next, we will discuss the role of the tumor microenvironment, focusing on interactions with stromal cells, dynamics of the extracellular matrix, and the involvement of immune cells [4][6]. Following this, we will examine the molecular pathways that regulate metastasis, highlighting key signaling networks and genetic and epigenetic regulations that contribute to metastatic behavior [2][6].
In the subsequent sections, we will review current therapeutic strategies and emerging targets for intervention in metastatic disease [1][7]. We will also consider future directions in metastasis research, including novel approaches and the importance of early detection in improving patient outcomes [1][6]. By synthesizing current knowledge in the field, this review aims to provide a comprehensive overview of the mechanisms underlying cancer metastasis and to identify critical areas for future research.
2 Mechanisms of Cancer Metastasis
2.1 Invasion of Cancer Cells
Cancer metastasis is a complex biological process that involves multiple mechanisms facilitating the spread of cancer cells from the primary tumor to distant organs. The invasion of cancer cells is a critical initial step in this metastatic cascade, characterized by several interrelated processes.
The metastatic process begins with the detachment of cancer cells from the primary tumor, which is often facilitated by the loss of cell adhesion molecules such as E-cadherin, a hallmark of epithelial-mesenchymal transition (EMT). EMT is crucial as it allows cancer cells to acquire migratory and invasive properties, enabling them to invade surrounding tissues and enter the bloodstream or lymphatic system [5].
Once detached, cancer cells undergo a series of changes that enhance their invasive capabilities. For instance, they can modify their cytoskeletal structures and express proteolytic enzymes, such as matrix metalloproteinases (MMPs), which degrade the extracellular matrix (ECM) and basement membranes, allowing for further invasion into adjacent tissues [8].
In addition to these cellular changes, the tumor microenvironment plays a significant role in promoting invasion. Various components within the microenvironment, including stromal cells, immune cells, and the ECM, interact with cancer cells to support their invasive behavior. For example, tumor-associated macrophages (TAMs) can secrete cytokines and growth factors that promote cancer cell migration and invasion [11]. Moreover, the ECM itself can influence cancer cell behavior; its stiffness and composition can dictate the migratory pathways of cancer cells and their ability to invade [5].
Epigenetic changes also contribute to the invasive characteristics of cancer cells. These changes can affect gene expression patterns, leading to the activation of pathways that promote invasion and migration. For instance, long non-coding RNAs (lncRNAs) have been shown to modulate signaling pathways that facilitate the dissemination of carcinoma cells [4].
Furthermore, the role of metabolic reprogramming in cancer invasion cannot be overlooked. Cancer cells often undergo metabolic changes that support their increased energy demands during invasion and migration. These metabolic alterations can influence various cellular processes, including the EMT and survival of cancer cells in circulation [8].
Overall, the invasion of cancer cells is a multifaceted process driven by a combination of cellular, molecular, and environmental factors. Understanding these mechanisms is crucial for developing targeted therapies aimed at inhibiting metastasis and improving patient outcomes. The intricate interplay between cancer cells and their microenvironment highlights the complexity of metastatic progression and underscores the need for continued research in this area [1][2].
2.2 Intravasation into Blood Vessels
Cancer metastasis is a complex and multistep process that involves the dissemination of cancer cells from the primary tumor to distant sites in the body, leading to the formation of secondary tumors. One of the critical steps in this metastatic cascade is intravasation, where cancer cells penetrate the endothelial barrier and enter the bloodstream. This process is influenced by various intrinsic and extrinsic factors.
Intravasation begins with the detachment of tumor cells from the primary tumor. This detachment is facilitated by changes in the tumor microenvironment, which can include the presence of specific growth factors and alterations in cell adhesion properties. For instance, the tumor microenvironment of metastasis (TMEM) has been identified as a key area where intravasation occurs, characterized by a unique interaction between tumor cells, macrophages, and endothelial cells. In this context, the Tie2high/VEGFhigh perivascular macrophages play a crucial role in promoting the intravasation of cancer cells by interacting with them and the endothelial cells, thereby facilitating their entry into the bloodstream[12].
Moreover, cancer cells employ various mechanisms to breach the endothelial barrier. For example, studies have shown that specific signaling pathways and receptor interactions are crucial for this process. Platelets have been identified as significant contributors to all steps of hematogenous tumor dissemination, including intravasation. They aid in tumor cell-induced aggregation and help protect circulating tumor cells from immune surveillance, thereby enhancing their chances of survival in the bloodstream[13].
The mechanical properties of the endothelial barrier also play a vital role in intravasation. Endothelial cells exhibit heterogeneity, and their mechanical characteristics can influence how easily cancer cells can penetrate them. Research indicates that factors such as vascular flow and the extracellular matrix (ECM) composition of the perivascular niche can significantly affect the efficiency of intravasation[14].
Additionally, metabolic factors, such as systemic cholesterol levels, have been shown to influence intravasation. A high-cholesterol diet has been linked to increased intravasation of breast tumor cells through mechanisms involving low-density lipoprotein (LDL) and its receptor, LDLR, which promote vascular invasion and alter the adhesion properties of tumor cells[15].
The process of intravasation is also influenced by various signaling molecules and cellular responses. For instance, epithelial-mesenchymal transition (EMT) is a critical process that enhances the invasive capabilities of cancer cells. During intravasation, cancer cells may undergo a switch in their expression profiles, characterized by an initial increase in mesenchymal markers such as Vimentin, followed by a decrease alongside an increase in epithelial markers like epithelial cell adhesion molecule (EpCAM)[16].
In summary, the mechanisms of cancer metastasis, particularly intravasation into blood vessels, involve a multifaceted interplay of cellular signaling, mechanical properties of the endothelial barrier, interactions with the tumor microenvironment, and metabolic influences. Understanding these mechanisms is crucial for developing therapeutic strategies aimed at preventing metastasis and improving patient outcomes.
2.3 Survival in Circulation
Cancer metastasis is a complex process that involves several critical steps, one of which is the survival of cancer cells in circulation. This phase is crucial because circulating tumor cells (CTCs) must endure various hostile conditions within the bloodstream before they can extravasate and establish secondary tumors. The survival mechanisms of CTCs are essential to understanding metastasis and developing effective therapeutic strategies.
Circulating tumor cells face significant challenges in the circulatory system, including hydrodynamic shear stress (HSS), oxidative damage, anoikis (a form of programmed cell death that occurs when cells detach from the extracellular matrix), and immune surveillance. These factors can severely limit the lifespan and viability of CTCs. Despite these challenges, a specific subset of CTCs has been shown to persist in the bloodstream, indicating the presence of survival mechanisms that allow them to withstand these adverse conditions [17].
One of the primary survival mechanisms involves the ability of CTCs to adapt to the hydrodynamic shear stress encountered in the bloodstream. Studies have demonstrated that CTCs can exhibit changes in their mechanical properties, allowing them to endure the forces exerted by blood flow. For instance, cancer cells that survive prolonged circulatory stress often show increased expression of epithelial-to-mesenchymal transition (EMT) markers, which are associated with enhanced migratory and invasive capabilities [18]. This EMT process is pivotal for the survival of CTCs as it enables them to acquire characteristics that facilitate their adaptation to the circulatory environment.
Additionally, the interactions between CTCs and the components of the tumor microenvironment (TME) play a significant role in their survival. The TME can provide supportive signals that enhance the resilience of CTCs, allowing them to evade immune detection and resist anoikis [19]. The interplay between tumor-derived metabolites and CTCs also influences their metastatic potential. Metabolites can modulate the expression of various genes involved in survival pathways, further reinforcing the CTCs' ability to persist in circulation [8].
Moreover, CTCs can exploit the physical properties of blood and lymphatic fluids to enhance their survival. For instance, the flow mechanics within these fluids can be manipulated by CTCs to optimize their transit and reduce the likelihood of being targeted by immune cells [20]. The dynamics of blood circulation and the interactions with endothelial cells are crucial for the CTCs' ability to lodge in capillaries and eventually extravasate into distant organs [21].
In summary, the survival of circulating tumor cells is governed by a combination of mechanical adaptations, interactions with the tumor microenvironment, and metabolic reprogramming. These mechanisms not only enable CTCs to withstand the hostile conditions of the bloodstream but also facilitate their eventual colonization of distant sites, making them a focal point in the study of cancer metastasis and the development of targeted therapies. Understanding these survival strategies is essential for devising interventions that could potentially disrupt the metastatic cascade and improve patient outcomes [22].
2.4 Extravasation and Colonization
Cancer metastasis is a complex, multistep process that significantly contributes to cancer-related mortality. Among the critical stages of metastasis are extravasation and colonization, both of which involve intricate molecular and cellular mechanisms.
Extravasation refers to the process by which circulating tumor cells (CTCs) exit the bloodstream and invade surrounding tissues at distant sites. This process is essential for the establishment of secondary tumors. During extravasation, cancer cells must first detach from the primary tumor, penetrate the endothelial barrier of blood vessels, and then migrate into the surrounding tissue. This step is often inefficient and transient, with numerous obstacles that cancer cells must overcome, including the need to evade immune responses and adapt to the new microenvironment [23].
The mechanisms underlying extravasation are not yet fully understood, but several factors have been identified that facilitate this process. For instance, the physical properties of the microenvironment, such as the mechanical characteristics of the extracellular matrix (ECM) and the vascular endothelial glycocalyx, play significant roles in the ability of cancer cells to extravasate [24]. Mechanical cues, including the stiffness of the ECM and the dynamics of vascular flow, can influence the adhesion and transmigration of cancer cells through the endothelium [25]. Furthermore, the expression of specific adhesion molecules and the activation of intracellular signaling pathways are critical for successful extravasation [26].
Once CTCs extravasate, they must establish themselves in the new tissue environment to form metastatic colonies. This colonization phase involves a series of additional challenges, including the need for cancer cells to survive and proliferate in an unfamiliar microenvironment. The metastatic colonization process is influenced by both intrinsic properties of the cancer cells and extrinsic factors from the host tissue [9]. The tumor microenvironment can significantly affect the behavior of disseminated tumor cells, as interactions with stromal cells, immune cells, and ECM components create a supportive niche for tumor growth [27].
Phenotypic plasticity of cancer cells is also a crucial determinant of metastatic colonization. This plasticity allows tumor cells to adapt to various microenvironments, enhancing their survival and proliferation capabilities [28]. Epithelial-mesenchymal transition (EMT) is one such mechanism that enables cancer cells to acquire migratory and invasive properties, facilitating their transition from the primary tumor to metastatic sites [5]. Conversely, once in the metastatic site, some cancer cells may undergo mesenchymal-epithelial transition (MET), allowing them to thrive in the new environment [29].
In summary, the mechanisms of cancer metastasis, particularly during the stages of extravasation and colonization, involve a combination of cellular behaviors, mechanical properties of the environment, and molecular signaling pathways. Understanding these mechanisms is critical for developing effective therapeutic strategies aimed at preventing and treating metastatic disease.
3 Role of the Tumor Microenvironment
3.1 Interactions with Stromal Cells
Cancer metastasis is a complex process that involves multiple mechanisms, with significant contributions from the tumor microenvironment (TME). The TME comprises various components, including stromal cells, extracellular matrix (ECM), immune cells, and signaling molecules, all of which interact dynamically with cancer cells. These interactions play crucial roles in promoting tumor progression, invasion, and metastasis.
One of the primary mechanisms by which the TME facilitates metastasis is through tumor-stromal interactions. Tumor cells engage with stromal cells, such as fibroblasts, macrophages, and endothelial cells, leading to reciprocal signaling that enhances tumor growth and metastatic potential. For instance, the interactions between cancer cells and the stroma regulate critical processes, including the degradation of the ECM, which is essential for tumor invasion and dissemination. The release of growth factors and cytokines from stromal cells can further activate pro-tumor signaling pathways in cancer cells, promoting their survival and migration [30].
MicroRNAs (miRNAs) also play a pivotal role in mediating interactions between cancer cells and stromal cells. These small RNA molecules can influence the expression of target genes involved in cancer cell proliferation, migration, and the remodeling of the microenvironment. By acting as regulatory messengers, secreted miRNAs can facilitate communication between cancer and stromal cells, thereby supporting the establishment of a metastatic niche [31].
Additionally, the tumor microenvironment can modulate the formation and function of invadopodia, which are actin-rich structures that cancer cells use to invade surrounding tissues. Signals from the TME, including growth factors and hypoxic conditions, can enhance the activity of invadopodia, thereby promoting cancer cell invasion and metastasis [32].
The stromal microenvironment's role is particularly pronounced in breast cancer metastasis, where a bidirectional interplay exists between breast cancer cells and stromal components. This interaction can activate signaling pathways that enhance metastatic spread, demonstrating that the TME is not merely a passive bystander but actively contributes to tumor progression [33].
Furthermore, the recruitment of stromal cells from the microenvironment is a critical step in the metastatic process. As tumor cells escape the primary site, they can induce changes in the surrounding stroma that support their survival and proliferation at distant sites. This includes the recruitment of immune cells and the secretion of chemokines that further modify the TME to favor metastasis [34].
Overall, the tumor microenvironment, through its complex interactions with stromal cells, plays a vital role in the mechanisms of cancer metastasis. Understanding these interactions provides insights into potential therapeutic targets aimed at disrupting the supportive roles of the TME in cancer progression. Targeting both cancer cells and their associated microenvironment may yield more effective strategies for preventing and treating metastatic disease [35][36].
3.2 Extracellular Matrix Dynamics
Cancer metastasis is a complex and multistep process that involves the dissemination of cancer cells from the primary tumor to distant sites, ultimately leading to the formation of secondary tumors. The tumor microenvironment, particularly the extracellular matrix (ECM), plays a pivotal role in this process by influencing cancer cell behavior, including migration, invasion, and survival.
The ECM provides structural support to tissues and serves as a dynamic interface between cancer cells and their surrounding environment. Alterations in the ECM can significantly affect cancer progression. For instance, ECM remodeling, which involves changes in its composition and mechanical properties, can facilitate tumor cell invasion and migration. Cancer cells can physically and chemically remodel the ECM, impacting critical cellular behaviors such as recognition of matrix geometry and rigidity, cell polarization, motility, and cytoskeletal reorganization [37].
Mechanotransduction, the process by which cells convert mechanical stimuli from their environment into biochemical signals, is crucial in cancer metastasis. The mechanical properties of the ECM, including stiffness and elasticity, can dictate how cancer cells respond to external forces. Migrating tumor cells exhibit increased motility as they traverse the ECM, which is influenced by the mechanical characteristics of the surrounding matrix [38].
The interaction between cancer cells and the ECM is mediated by various signaling pathways. For example, integrins, which are transmembrane receptors, play a critical role in the adhesion of cancer cells to the ECM. This adhesion not only facilitates cell migration but also activates intracellular signaling cascades that promote survival and proliferation [39]. Additionally, the ECM can secrete signaling molecules that enhance cancer cell invasiveness and modulate the immune response, creating a supportive niche for metastasis [40].
Inflammation within the tumor microenvironment also contributes to ECM dynamics and cancer metastasis. Pro-inflammatory cytokines can alter ECM composition and promote a permissive environment for cancer cell migration. The interplay between inflammatory signals and ECM remodeling is essential for understanding the mechanisms driving metastasis [41].
Furthermore, the mechanical properties of the ECM can influence the behavior of circulating tumor cells (CTCs). As CTCs travel through the bloodstream, they encounter various physical barriers, including the endothelial glycocalyx and the subendothelial ECM, which can impact their ability to extravasate and establish secondary tumors [24]. The physical characteristics of the metastatic microenvironment, including its stiffness and the presence of specific ECM components, can dictate the fate of CTCs and their potential to colonize distant organs [42].
In summary, the dynamics of the tumor microenvironment, particularly the ECM, are integral to the mechanisms of cancer metastasis. By influencing cellular behaviors through mechanical and biochemical cues, the ECM not only facilitates the migration and invasion of cancer cells but also contributes to the establishment of a supportive metastatic niche. Understanding these interactions provides insights into potential therapeutic strategies aimed at disrupting the metastatic process and targeting the tumor microenvironment to inhibit cancer progression [38][43][44].
3.3 Immune Cell Involvement
Cancer metastasis is a complex process characterized by the spread of cancer cells from the primary tumor to distant organs, significantly contributing to cancer-related mortality. The mechanisms underlying this phenomenon are intricately linked to the tumor microenvironment, particularly the involvement of various immune cells.
The tumor microenvironment plays a pivotal role in facilitating metastasis. It consists of a diverse array of cellular and non-cellular components, including immune cells, stromal cells, extracellular matrix, and signaling molecules. These components interact dynamically with cancer cells, influencing their invasive capabilities and survival during metastasis. Specifically, the interactions between tumor cells and immune cells can either promote or inhibit metastatic processes.
Innate immune cells, such as tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and natural killer (NK) cells, are particularly influential in the metastatic cascade. TAMs are known to promote tumor growth, invasion, and metastasis by creating a supportive microenvironment through the secretion of cytokines and growth factors that remodel the extracellular matrix and enhance tumor cell motility. They can also facilitate angiogenesis, which is crucial for supplying nutrients to the growing tumor and aiding its dissemination (Smith & Kang 2013) [45].
MDSCs are another critical component of the tumor microenvironment, known for their immunosuppressive functions. They can inhibit T cell responses and promote tumor progression, thereby facilitating metastasis. Their role in the tumor microenvironment is to create conditions that allow tumor cells to evade immune surveillance and establish secondary tumors (Yin et al. 2019) [46].
Furthermore, the lymph node microenvironment has been identified as a significant site for metastasis. It serves as a first metastatic niche for various cancers, where immune cells interact closely with cancer cells. This interaction can enhance the metastatic potential of tumor cells through epigenetic reprogramming, which may lead to increased malignancy (Li et al. 2022) [3].
In addition to direct interactions, the immune microenvironment also contributes to the formation of a premetastatic niche in distant organs. This niche is established by the recruitment of immune cells that modify the local environment, making it more conducive for tumor cell colonization (Gao et al. 2023) [47]. For instance, the presence of pro-inflammatory cytokines and chemokines secreted by immune cells can enhance the invasive characteristics of cancer cells, facilitating their extravasation and colonization in new tissues (Wu & Zhou 2009) [41].
Moreover, inflammation within the tumor microenvironment has been recognized as a driving force in cancer metastasis. The inflammatory milieu, characterized by the presence of infiltrating immune cells and their secreted factors, significantly contributes to the invasive and metastatic traits of cancer cells (Bogenrieder & Herlyn 2003) [48].
In summary, the mechanisms of cancer metastasis are profoundly influenced by the tumor microenvironment, particularly through the involvement of immune cells. These cells not only interact with tumor cells to promote invasion and survival but also modify the local environment to facilitate metastatic spread. Understanding these interactions is crucial for developing effective therapeutic strategies aimed at inhibiting metastasis and improving cancer outcomes.
4 Molecular Pathways in Metastasis
4.1 Key Signaling Pathways
Cancer metastasis is a complex and multi-step process that involves the dissemination of malignant cells from a primary tumor to secondary sites, leading to the establishment of secondary tumors. Understanding the molecular mechanisms underlying this process is crucial for developing effective therapeutic strategies. Key signaling pathways that have been implicated in cancer metastasis include the Hippo pathway, mitogen-activated protein kinase (MAPK) pathways, and calcium signaling, among others.
The Hippo pathway is an emerging signaling cascade that plays significant roles in tumor initiation and progression. Dysregulation of the Hippo pathway has been shown to promote various aspects of cancer metastasis. This pathway's core components are involved in regulating cell proliferation and apoptosis, which are critical in the metastatic cascade. Recent studies have provided clinical and biochemical evidence linking the Hippo pathway to cancer metastasis, highlighting its potential as a target for therapeutic intervention (Janse van Rensburg and Yang, 2016) [10].
The MAPK signaling pathways, including ERK, JNK, and P38, are also crucial in modulating cancer cell motility and metastasis. Activation of the ERK MAPK pathway has been associated with enhanced cell motility, which is a key component of the metastatic process. This pathway regulates various cellular functions that facilitate the dissemination of cancer cells from the primary tumor (Viala and Pouysségur, 2004) [49]. Additionally, the MAPK pathways are involved in the coordination of signaling events that promote the survival of cancer cells in circulation and their adaptation to foreign microenvironments upon extravasation (Kciuk et al., 2022) [50].
Calcium (Ca2+) signaling has emerged as another important player in cancer metastasis. Ca2+ acts as a ubiquitous secondary messenger, and its dysregulation is linked to various processes associated with tumor progression, including cellular adhesion, epithelial-mesenchymal transition (EMT), and cell migration. Spatiotemporal regulation of Ca2+ homeostasis is critical for tumor progression and metastasis, suggesting that modulation of Ca2+ signaling pathways may offer new therapeutic avenues for treating metastatic cancers (Alharbi et al., 2021) [51].
Moreover, metabolic reprogramming in cancer cells is closely associated with the metastatic process. Cancer cells often undergo metabolic changes that facilitate their survival and proliferation in distant organs. This metabolic rewiring influences various stages of metastasis, including EMT, circulation survival, and colonization at secondary sites. Understanding the interplay between metabolic pathways and metastatic signaling can reveal novel molecular targets for intervention (Wei et al., 2020) [8].
In addition to these pathways, the interaction between tumor cells and the tumor microenvironment is pivotal in the metastatic process. The "seed and soil" hypothesis emphasizes the importance of permissive interactions between tumor cells and the microenvironments of secondary organs. Growth factor signaling pathways have been shown to play dual roles in metastasis, acting both as promoters and inhibitors depending on the context (Lowery and Yu, 2012) [52].
Overall, the mechanisms of cancer metastasis involve a complex network of signaling pathways and interactions with the tumor microenvironment. Continued research into these pathways will enhance our understanding of metastasis and contribute to the development of targeted therapies aimed at mitigating this lethal aspect of cancer progression.
4.2 Genetic and Epigenetic Regulation
Cancer metastasis is a complex multistep process that involves the dissemination of cancer cells from the primary tumor to distant sites, leading to the formation of secondary tumors. This process is regulated by a variety of genetic and epigenetic mechanisms that influence tumor initiation, growth, and the metastatic cascade.
Genetic alterations, such as mutations, amplifications, and translocations in oncogenes (e.g., RAS and MYC) and tumor suppressor genes (e.g., TP53 and PTEN), play a significant role in cancer progression and metastasis. These genetic changes can lead to genomic instability, which contributes to tumor heterogeneity and clonal evolution, thereby enhancing the metastatic potential of cancer cells [53].
In addition to genetic factors, epigenetic modifications are crucial in regulating cancer metastasis. Epigenetic changes, including DNA methylation, histone modifications, and the regulation by non-coding RNAs (ncRNAs), significantly influence gene expression without altering the underlying DNA sequence. Long non-coding RNAs (lncRNAs) are particularly important epigenetic regulators that modulate various signaling pathways involved in metastasis. They act as decoys, guides, and scaffolds, impacting key molecules at different stages of the metastatic process, including cell dissemination, intravascular transit, and colonization of distant organs [4][54].
The epithelial to mesenchymal transition (EMT) is a critical process in metastasis, where cancer cells lose their epithelial characteristics and gain migratory and invasive properties. This transition is regulated by a network of genetic and epigenetic factors that include lncRNAs, microRNAs (miRNAs), and chromatin remodeling [55][56]. For instance, miRNAs have been shown to regulate metastatic gene pathways, influencing processes such as cell migration and invasion [6].
Moreover, the tumor microenvironment plays a vital role in supporting metastasis. Interactions between cancer cells and the surrounding stromal cells can facilitate the metastatic process through mechanisms such as immune evasion, angiogenesis, and the remodeling of the extracellular matrix [48]. Epigenetic alterations in both cancer cells and the stromal microenvironment can further enhance the metastatic potential [57].
Recent advances in technologies such as CRISPR-Cas9 have enabled researchers to investigate the specific roles of epigenetic modifications in metastasis, revealing how these changes can drive tumor progression and therapy resistance [57]. Understanding these intricate molecular pathways is essential for developing targeted therapies aimed at inhibiting metastasis and improving clinical outcomes for cancer patients.
In summary, the mechanisms of cancer metastasis involve a dynamic interplay of genetic mutations and epigenetic modifications, along with the influence of the tumor microenvironment. Continued research into these mechanisms is crucial for identifying novel therapeutic strategies to combat metastatic disease.
5 Therapeutic Implications
5.1 Current Treatment Strategies
Cancer metastasis is a complex multistep process characterized by the dissemination of cancer cells from the primary tumor to distant organs, ultimately leading to secondary tumor formation. The mechanisms underlying this process involve a series of biological events, including but not limited to, invasion of adjacent tissues, intravasation into the bloodstream, transport through the circulatory system, arrest at a secondary site, extravasation from blood vessels, and growth in the new organ environment. Each of these steps is influenced by the tumor's ability to induce angiogenesis, thus facilitating its own blood supply, which is crucial for sustaining tumor growth and metastasis [58].
The metastatic process is significantly affected by the tumor microenvironment, which plays a vital role in facilitating the invasion and proliferation of cancer cells. The interactions between tumor cells and their microenvironment can alter cellular signaling pathways and metabolic processes, thereby enhancing metastatic potential [59]. For instance, specific metabolic patterns can favor the adaptation of cancer cells to foreign microenvironments, contributing to the establishment of secondary tumors [60]. Furthermore, mechanisms such as epithelial-mesenchymal transition (EMT) and resistance to anoikis (a form of programmed cell death) are critical for enabling cancer cells to survive and thrive in new locations [1].
Current treatment strategies for metastatic cancer primarily focus on conventional therapies, including surgery, chemotherapy, and radiotherapy. However, these approaches have limitations, particularly concerning their efficacy against metastatic disease. Recent advancements have introduced novel therapeutic strategies targeting specific aspects of the metastatic process. For instance, agents that inhibit tumor invasion, angiogenesis, and specific signaling pathways are under development. Targeting cancer stem cells, which are believed to play a pivotal role in orchestrating the metastatic niche, is also a promising area of research [61].
The integration of targeted therapies and immunotherapies represents a significant shift in the management of metastatic cancer. These approaches aim to disrupt the molecular pathways that facilitate metastasis, thereby improving patient outcomes. For example, the targeting of transcription factors such as FOXC1, which is implicated in driving cancer metastasis, offers a potential therapeutic avenue [62]. Moreover, understanding the genetic and epigenetic changes in metastatic cells may lead to the identification of novel biomarkers that can predict metastatic risk and inform treatment strategies [63].
In conclusion, while significant progress has been made in understanding the mechanisms of cancer metastasis, challenges remain in translating this knowledge into effective treatments. The complexity of the metastatic cascade necessitates a multifaceted approach, combining traditional therapies with innovative strategies that target the underlying biological mechanisms of metastasis. Continued research into the molecular and cellular interactions within the tumor microenvironment will be crucial for developing more effective therapeutic interventions against metastatic cancer [64].
5.2 Emerging Therapeutic Targets
Cancer metastasis is a complex and multistep process involving the spread of cancer cells from the primary tumor to distant sites, significantly contributing to cancer-related morbidity and mortality. The mechanisms underlying this process encompass a series of biological events, including invasion, intravasation, circulation, extravasation, and colonization at secondary sites. Each of these steps is influenced by various cellular and molecular factors, as well as interactions with the tumor microenvironment.
The initial step in metastasis is the invasion of adjacent tissues, where cancer cells breach the surrounding extracellular matrix (ECM) and migrate into adjacent tissues. This process is often facilitated by the epithelial-mesenchymal transition (EMT), a biological process where epithelial cells lose their cell polarity and adhesion properties, gaining migratory and invasive capabilities. The tumor microenvironment plays a critical role in this transition, as factors such as growth factors, cytokines, and extracellular matrix components can promote or inhibit these processes [1].
Following invasion, cancer cells enter the bloodstream or lymphatic system, a process termed intravasation. Here, they must survive in circulation, evading immune surveillance and the shear stress of blood flow. This survival is often aided by the formation of circulating tumor cell (CTC) clusters, which exhibit enhanced survival compared to single CTCs [6]. Once in the circulation, the cells can disseminate to distant organs, where they can arrest at specific sites—a phenomenon influenced by the organ-specific tropism of certain cancer types [65].
Upon reaching a secondary site, cancer cells undergo extravasation, wherein they exit the blood vessels and invade the surrounding tissue. This step is followed by colonization, where the metastatic cells adapt to the new microenvironment, often referred to as the metastatic niche. The ability of disseminated cancer cells to survive and proliferate in these new environments is influenced by various factors, including the availability of nutrients, the presence of supportive stromal cells, and the immune landscape of the host [2].
The therapeutic implications of understanding these mechanisms are profound. Recent research has focused on identifying emerging therapeutic targets that can disrupt various stages of the metastatic cascade. For instance, targeting the EMT process and the signaling pathways involved in cell migration and invasion can potentially hinder the initial steps of metastasis [66]. Furthermore, therapies aimed at modulating the tumor microenvironment, such as immunotherapies that enhance anti-tumor immune responses or agents that disrupt the supportive stroma, have shown promise in preclinical and clinical settings [67].
Moreover, the role of microRNAs (miRNAs) in regulating metastatic pathways has emerged as a significant area of interest. Aberrant expression of specific miRNAs has been implicated in the regulation of genes associated with metastasis, suggesting that miRNA-based therapies could offer novel strategies for combating metastatic disease [6].
Overall, the intricate interplay between cancer cells and their microenvironment underscores the necessity for a multifaceted approach to therapy, combining traditional modalities with innovative strategies targeting the metastatic process at various levels. The identification of specific molecular pathways and the development of agents that can effectively inhibit these pathways hold the potential to significantly improve outcomes for patients with metastatic cancer [1][68].
6 Future Directions in Metastasis Research
6.1 Novel Approaches and Technologies
Cancer metastasis is a complex and multifaceted process that involves the dissemination of cancer cells from the primary tumor to distant organs, leading to secondary tumor formation. Understanding the mechanisms of metastasis is crucial for developing effective therapeutic strategies. The following outlines the key mechanisms and highlights future directions in metastasis research.
Epithelial-Mesenchymal Transition (EMT): EMT is a critical step in metastasis where epithelial cells acquire mesenchymal traits, enhancing their migratory and invasive capabilities. This transition is regulated by various signaling pathways, including TGF-β, Wnt, and Notch pathways. EMT allows cancer cells to detach from the primary tumor, invade surrounding tissues, and enter the bloodstream or lymphatic system [4].
Anoikis Resistance: Cancer cells often develop resistance to anoikis, a form of programmed cell death that occurs when cells detach from the extracellular matrix. This resistance enables them to survive in circulation and establish metastatic colonies in distant sites [2].
Tumor Microenvironment (TME): The TME plays a significant role in supporting metastasis. Components such as extracellular matrix (ECM), stromal cells, and immune cells interact with cancer cells, influencing their behavior. For instance, the presence of immune cells can either promote or inhibit metastasis, depending on the context [3].
Metabolic Reprogramming: Cancer cells often undergo metabolic changes that support their survival and proliferation during metastasis. For example, the Warburg effect, where cancer cells preferentially utilize glycolysis even in the presence of oxygen, provides them with the necessary energy and biosynthetic precursors to thrive in distant microenvironments [69].
Mechanical Properties and Homeostasis: The mechanical properties of both cancer cells and their surrounding environment can influence metastasis. Changes in ECM stiffness and cellular mechanical stress can promote EMT and facilitate tumor cell migration [5].
Genetic and Epigenetic Alterations: Genetic mutations and epigenetic changes can drive the metastatic process. Long non-coding RNAs (lncRNAs) and microRNAs (miRNAs) have been identified as critical regulators of metastasis, modulating various signaling pathways and gene expressions involved in cancer progression [4][70].
Cellular Interactions: The interaction between cancer cells and surrounding stromal cells is crucial for metastasis. For instance, gap junctions can facilitate communication between tumor cells and stromal cells, influencing cell adhesion and migration [71].
Future directions in metastasis research include:
Targeting the Tumor Microenvironment: Developing therapeutic strategies that focus on reprogramming the TME to inhibit metastatic progression could yield significant clinical benefits. This may involve targeting specific cell types within the TME or modulating ECM components [62].
Understanding Metabolic Mechanisms: Investigating the metabolic adaptations of cancer cells during metastasis could lead to novel therapeutic targets. Inhibitors of metabolic pathways that support metastatic growth may prove effective [8].
Advanced Technologies: Utilizing technologies such as single-cell sequencing and advanced imaging techniques can provide deeper insights into the heterogeneity of metastatic tumors and the dynamics of cell interactions within the TME [9].
Biomarker Development: Identifying biomarkers that predict metastatic potential at early stages of cancer could facilitate timely intervention and improve patient outcomes. Biomarkers related to EMT, metabolic reprogramming, and immune evasion are particularly promising [62].
In conclusion, the mechanisms of cancer metastasis are intricate and involve a multitude of biological processes. Continued research in this field, particularly focusing on the interplay between cancer cells and their microenvironment, will be essential for developing effective therapies against metastatic disease.
6.2 Importance of Early Detection
Cancer metastasis is a multifaceted biological process characterized by the dissemination of cancer cells from a primary tumor to distant sites in the body, which ultimately leads to the majority of cancer-related deaths. Understanding the mechanisms of metastasis is crucial for developing effective therapeutic strategies. The following are key mechanisms involved in cancer metastasis:
Epithelial-Mesenchymal Transition (EMT): EMT is a critical process that enables epithelial cancer cells to acquire mesenchymal characteristics, facilitating their invasion and migration. This transition involves the downregulation of epithelial markers like E-cadherin and the upregulation of mesenchymal markers such as N-cadherin and vimentin, promoting increased motility and invasiveness of cancer cells (Liu et al. 2021; Fan et al. 2022).
Tumor Microenvironment (TME): The TME plays a significant role in supporting metastatic processes. Components such as extracellular matrix (ECM), immune cells, and stromal cells interact dynamically with cancer cells, influencing their behavior and enhancing metastatic potential. For instance, the presence of tumor-associated macrophages and fibroblasts can promote tumor cell survival, migration, and invasion (Li et al. 2022; Suresh and Guruvayoorappan 2023).
Genetic and Epigenetic Changes: Cancer cells often undergo genetic mutations and epigenetic modifications that enhance their metastatic capabilities. These alterations can affect various signaling pathways involved in cell adhesion, migration, and invasion. Long non-coding RNAs (lncRNAs) have emerged as important regulators of these processes, modulating the expression of genes involved in metastasis (Khurshid et al. 2023).
Cell Adhesion and Migration: The metastatic cascade involves a series of steps including detachment from the primary tumor, migration through the ECM, intravasation into blood or lymphatic vessels, survival in circulation, and extravasation into distant tissues. Cell adhesion molecules such as integrins and cadherins play pivotal roles in mediating these interactions (Bashyam 2002; Janse van Rensburg and Yang 2016).
Metabolic Reprogramming: Cancer cells often exhibit altered metabolism that supports their survival and proliferation in distant sites. This metabolic rewiring can influence the metastatic process by enhancing cell survival during circulation and promoting the colonization of metastatic sites (Wei et al. 2020).
Immune Evasion: Metastatic cancer cells can evade immune surveillance through various mechanisms, including the secretion of immunosuppressive factors and the alteration of immune cell recruitment. This immune evasion facilitates their survival and growth in new environments (Xiong et al. 2022).
Angiogenesis: The formation of new blood vessels (angiogenesis) is crucial for providing nutrients and oxygen to metastatic tumors. Tumor cells can secrete pro-angiogenic factors that stimulate vascular growth, supporting their metastatic growth (Flamini et al. 2016).
Pre-Metastatic Niche Formation: Before the arrival of cancer cells, certain organs can undergo changes that create a favorable environment for metastasis. This pre-metastatic niche is established through the recruitment of immune cells and the secretion of factors that prepare the tissue for incoming tumor cells (Li et al. 2022).
Future directions in metastasis research should focus on elucidating these complex mechanisms in greater detail, with an emphasis on identifying novel therapeutic targets and developing strategies for early detection. The integration of advanced technologies such as single-cell sequencing and imaging techniques can provide deeper insights into the cellular and molecular dynamics of metastasis. Furthermore, understanding the role of lifestyle factors and environmental influences on metastatic processes could lead to innovative prevention strategies.
The importance of early detection in cancer metastasis cannot be overstated. Early identification of metastatic disease significantly improves treatment outcomes and survival rates. Developing reliable biomarkers and screening methods for the early detection of metastasis will be crucial in reducing cancer mortality and enhancing patient prognosis (Guan 2015).
In summary, the mechanisms of cancer metastasis are complex and involve a variety of cellular, molecular, and environmental factors. Ongoing research is essential to uncover these mechanisms and translate findings into clinical applications for improving cancer management and patient care.
7 Conclusion
The investigation into the mechanisms of cancer metastasis has revealed several critical insights into the biological processes that underlie the spread of cancer cells from primary tumors to distant sites. Key findings emphasize the role of epithelial-mesenchymal transition (EMT), which facilitates the invasive and migratory capabilities of cancer cells, alongside the tumor microenvironment's significant influence on these processes. The interplay between cancer cells and stromal components, immune cells, and extracellular matrix dynamics is crucial in promoting metastasis. Moreover, genetic and epigenetic alterations, including the involvement of long non-coding RNAs and microRNAs, highlight the complexity of regulatory networks that govern metastatic behavior. Future research directions should focus on developing targeted therapies that disrupt these pathways, exploring metabolic reprogramming in cancer cells, and enhancing early detection methods to improve patient outcomes. Understanding these multifaceted mechanisms will be vital in combating metastatic disease and reducing cancer-related mortality.
References
- [1] Mengmeng Liu;Jing Yang;Bushu Xu;Xing Zhang. Tumor metastasis: Mechanistic insights and therapeutic interventions.. MedComm(IF=10.7). 2021. PMID:34977870. DOI: 10.1002/mco2.100.
- [2] Dhanisha Sulekha Suresh;Chandrasekharan Guruvayoorappan. Molecular principles of tissue invasion and metastasis.. American journal of physiology. Cell physiology(IF=4.7). 2023. PMID:36939199. DOI: 10.1152/ajpcell.00348.2022.
- [3] Tianhang Li;Tianyao Liu;Zihan Zhao;Xinyan Xu;Shoubin Zhan;Shengkai Zhou;Ning Jiang;Wenjie Zhu;Rui Sun;Fayun Wei;Baofu Feng;Hongqian Guo;Rong Yang. The Lymph Node Microenvironment May Invigorate Cancer Cells With Enhanced Metastatic Capacities.. Frontiers in oncology(IF=3.3). 2022. PMID:35295999. DOI: 10.3389/fonc.2022.816506.
- [4] Sana Khurshid Baba;Sadaf Khursheed Baba;Rashid Mir;Imadeldin Elfaki;Naseh Algehainy;Mohammad Fahad Ullah;Jameel Barnawi;Faisal H Altemani;Mohammad Alanazi;Syed Khalid Mustafa;Tariq Masoodi;Ammira S Alshabeeb Akil;Ajaz A Bhat;Muzafar A Macha. Long non-coding RNAs modulate tumor microenvironment to promote metastasis: novel avenue for therapeutic intervention.. Frontiers in cell and developmental biology(IF=4.3). 2023. PMID:37384249. DOI: 10.3389/fcell.2023.1164301.
- [5] Se Jik Han;Sangwoo Kwon;Kyung Sook Kim. Contribution of mechanical homeostasis to epithelial-mesenchymal transition.. Cellular oncology (Dordrecht, Netherlands)(IF=4.8). 2022. PMID:36149601. DOI: 10.1007/s13402-022-00720-6.
- [6] Mohammad Alam Jafri;Mohammed Hussein Al-Qahtani;Jerry William Shay. Role of miRNAs in human cancer metastasis: Implications for therapeutic intervention.. Seminars in cancer biology(IF=15.7). 2017. PMID:28188828. DOI: 10.1016/j.semcancer.2017.02.004.
- [7] Xiangming Guan. Cancer metastases: challenges and opportunities.. Acta pharmaceutica Sinica. B(IF=14.6). 2015. PMID:26579471. DOI: 10.1016/j.apsb.2015.07.005.
- [8] Qinyao Wei;Yun Qian;Jun Yu;Chi Chun Wong. Metabolic rewiring in the promotion of cancer metastasis: mechanisms and therapeutic implications.. Oncogene(IF=7.3). 2020. PMID:32839493. DOI: 10.1038/s41388-020-01432-7.
- [9] Arthur W Lambert;Yun Zhang;Robert A Weinberg. Cell-intrinsic and microenvironmental determinants of metastatic colonization.. Nature cell biology(IF=19.1). 2024. PMID:38714854. DOI: 10.1038/s41556-024-01409-8.
- [10] Helena J Janse van Rensburg;Xiaolong Yang. The roles of the Hippo pathway in cancer metastasis.. Cellular signalling(IF=3.7). 2016. PMID:27519476. DOI: .
- [11] Kaifen Xiong;Min Qi;Tobias Stoeger;Jianglin Zhang;Shanze Chen. The role of tumor-associated macrophages and soluble mediators in pulmonary metastatic melanoma.. Frontiers in immunology(IF=5.9). 2022. PMID:36131942. DOI: 10.3389/fimmu.2022.1000927.
- [12] Lucia Borriello;George S Karagiannis;Camille L Duran;Anouchka Coste;Maja H Oktay;David Entenberg;John S Condeelis. The role of the tumor microenvironment in tumor cell intravasation and dissemination.. European journal of cell biology(IF=4.3). 2020. PMID:32800278. DOI: 10.1016/j.ejcb.2020.151098.
- [13] David Stegner;Sebastian Dütting;Bernhard Nieswandt. Mechanistic explanation for platelet contribution to cancer metastasis.. Thrombosis research(IF=3.4). 2014. PMID:24862136. DOI: .
- [14] Katharine R Bittner;Juan M Jiménez;Shelly R Peyton. Vascularized Biomaterials to Study Cancer Metastasis.. Advanced healthcare materials(IF=9.6). 2020. PMID:31977160. DOI: 10.1002/adhm.201901459.
- [15] Ana Magalhães;Vanessa Cesário;Diogo Coutinho;Inês Matias;Germana Domingues;Catarina Pinheiro;Teresa Serafim;Sérgio Dias. A high-cholesterol diet promotes the intravasation of breast tumor cells through an LDL-LDLR axis.. Scientific reports(IF=3.9). 2024. PMID:38658568. DOI: 10.1038/s41598-024-59845-3.
- [16] Fengtao Jiang;Yingqi Zhang;Guocheng Fang;Yao Wang;Alexander Dupuy;Jasmine Jin;Yi Shen;Khoon S Lim;Yinyan Wang;Yu Shrike Zhang;Ann-Na Cho;Hongxu Lu;Lining Arnold Ju. Intravasation-On-µDevice (INVADE): Engineering Dynamic Vascular Interfaces to Study Cancer Cell Intravasation.. Advanced materials (Deerfield Beach, Fla.)(IF=26.8). 2025. PMID:40223399. DOI: 10.1002/adma.202501466.
- [17] Shuang Zhou;Huanji Xu;Yichun Duan;Qiulin Tang;Huixi Huang;Feng Bi. Survival mechanisms of circulating tumor cells and their implications for cancer treatment.. Cancer metastasis reviews(IF=8.7). 2024. PMID:38436892. DOI: 10.1007/s10555-024-10178-7.
- [18] Keila Alvarado-Estrada;Lina Marenco-Hillembrand;Sushila Maharjan;Valerio Luca Mainardi;Yu Shrike Zhang;Natanael Zarco;Paula Schiapparelli;Hugo Guerrero-Cazares;Rachel Sarabia-Estrada;Alfredo Quinones-Hinojosa;Kaisorn L Chaichana. Circulatory shear stress induces molecular changes and side population enrichment in primary tumor-derived lung cancer cells with higher metastatic potential.. Scientific reports(IF=3.9). 2021. PMID:33531664. DOI: 10.1038/s41598-021-82634-1.
- [19] Agnieszka Swierczak;Jeffrey W Pollard. Myeloid Cells in Metastasis.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2020. PMID:31548218. DOI: 10.1101/cshperspect.a038026.
- [20] Gautier Follain;David Herrmann;Sébastien Harlepp;Vincent Hyenne;Naël Osmani;Sean C Warren;Paul Timpson;Jacky G Goetz. Fluids and their mechanics in tumour transit: shaping metastasis.. Nature reviews. Cancer(IF=66.8). 2020. PMID:31780785. DOI: 10.1038/s41568-019-0221-x.
- [21] Yusheng Lu;Shu Lian;Yunlong Cheng;Yuying Ye;Xiaodong Xie;Chengbin Fu;Chen Zhang;Yewei Zhu;M Iqbal Parker;Lee Jia. Circulation patterns and seed-soil compatibility factors cooperate to cause cancer organ-specific metastasis.. Experimental cell research(IF=3.5). 2019. PMID:30578764. DOI: 10.1016/j.yexcr.2018.12.015.
- [22] Ziyi Wang;Zehui Li;Xiangyu Sun;Wanfu Men;Yan Xu. Cellular components of tumor microenvironment: understanding their role in lymphatic metastasis of tumors.. Frontiers in pharmacology(IF=4.8). 2024. PMID:39726782. DOI: 10.3389/fphar.2024.1463538.
- [23] Ana Sofia Azevedo;Gautier Follain;Shankar Patthabhiraman;Sébastien Harlepp;Jacky G Goetz. Metastasis of circulating tumor cells: favorable soil or suitable biomechanics, or both?. Cell adhesion & migration(IF=3.5). 2015. PMID:26312653. DOI: 10.1080/19336918.2015.1059563.
- [24] Chinedu C Okorafor;Sanjana Shastri;Ke Wen;Eno E Ebong. Mechanisms of triple-negative breast cancer extravasation: Impact of the physical environment and endothelial glycocalyx.. FASEB journal : official publication of the Federation of American Societies for Experimental Biology(IF=4.2). 2024. PMID:38949120. DOI: 10.1096/fj.202400380R.
- [25] Baptiste Hamelin;Milan M S Obradović;Atul Sethi;Michal Kloc;Simone Münst;Christian Beisel;Katja Eschbach;Hubertus Kohler;Savas Soysal;Marcus Vetter;Walter P Weber;Michael B Stadler;Mohamed Bentires-Alj. Single-cell Analysis Reveals Inter- and Intratumour Heterogeneity in Metastatic Breast Cancer.. Journal of mammary gland biology and neoplasia(IF=3.6). 2023. PMID:38066300. DOI: 10.1007/s10911-023-09551-z.
- [26] Fayth L Miles;Freddie L Pruitt;Kenneth L van Golen;Carlton R Cooper. Stepping out of the flow: capillary extravasation in cancer metastasis.. Clinical & experimental metastasis(IF=3.2). 2008. PMID:17906932. DOI: 10.1007/s10585-007-9098-2.
- [27] Emma Nolan;Yibin Kang;Ilaria Malanchi. Mechanisms of Organ-Specific Metastasis of Breast Cancer.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2023. PMID:36987584. DOI: 10.1101/cshperspect.a041326.
- [28] Charly Jehanno;Milica Vulin;Veronica Richina;Federica Richina;Mohamed Bentires-Alj. Phenotypic plasticity during metastatic colonization.. Trends in cell biology(IF=18.1). 2022. PMID:35484037. DOI: 10.1016/j.tcb.2022.03.007.
- [29] Michelle B Chen;Yousef Javanmardi;Somayeh Shahreza;Bianca Serwinski;Amir Aref;Boris Djordjevic;Emad Moeendarbary. Mechanobiology in oncology: basic concepts and clinical prospects.. Frontiers in cell and developmental biology(IF=4.3). 2023. PMID:38020912. DOI: 10.3389/fcell.2023.1239749.
- [30] Kalyan C Nannuru;Rakesh K Singh. Tumor-stromal interactions in bone metastasis.. Current osteoporosis reports(IF=5.3). 2010. PMID:20425618. DOI: 10.1007/s11914-010-0011-6.
- [31] Yinghan Su;Xiaoya Li;Weidan Ji;Bin Sun;Can Xu;Zhaoshen Li;Guojun Qian;Changqing Su. Small molecule with big role: MicroRNAs in cancer metastatic microenvironments.. Cancer letters(IF=10.1). 2014. PMID:24184826. DOI: .
- [32] Christine M Gould;Sara A Courtneidge. Regulation of invadopodia by the tumor microenvironment.. Cell adhesion & migration(IF=3.5). 2014. PMID:24714597. DOI: 10.4161/cam.28346.
- [33] Lucas E L Terceiro;Chidalu A Edechi;Nnamdi M Ikeogu;Barbara E Nickel;Sabine Hombach-Klonisch;Tanveer Sharif;Etienne Leygue;Yvonne Myal. The Breast Tumor Microenvironment: A Key Player in Metastatic Spread.. Cancers(IF=4.4). 2021. PMID:34638283. DOI: 10.3390/cancers13194798.
- [34] Zahraa I Khamis;Ziad J Sahab;Qing-Xiang Amy Sang. Active roles of tumor stroma in breast cancer metastasis.. International journal of breast cancer(IF=3.0). 2012. PMID:22482059. DOI: 10.1155/2012/574025.
- [35] Leland W K Chung;Adam Baseman;Vasily Assikis;Haiyen E Zhau. Molecular insights into prostate cancer progression: the missing link of tumor microenvironment.. The Journal of urology(IF=6.8). 2005. PMID:15592017. DOI: 10.1097/01.ju.0000141582.15218.10.
- [36] Wan Zhang;Peng Huang. Cancer-stromal interactions: role in cell survival, metabolism and drug sensitivity.. Cancer biology & therapy(IF=4.6). 2011. PMID:21191189. DOI: 10.4161/cbt.11.2.14623.
- [37] GeonHui Lee;Seong-Beom Han;Jung-Hwan Lee;Hae-Won Kim;Dong-Hwee Kim. Cancer Mechanobiology: Microenvironmental Sensing and Metastasis.. ACS biomaterials science & engineering(IF=5.5). 2019. PMID:33405888. DOI: 10.1021/acsbiomaterials.8b01230.
- [38] Dimitra Vasilaki;Athina Bakopoulou;Alexandros Tsouknidas;Elaine Johnstone;Konstantinos Michalakis. Biophysical interactions between components of the tumor microenvironment promote metastasis.. Biophysical reviews(IF=3.7). 2021. PMID:34168685. DOI: 10.1007/s12551-021-00811-y.
- [39] J Thomas Parsons;Jill Slack-Davis;Robert Tilghman;W Gregory Roberts. Focal adhesion kinase: targeting adhesion signaling pathways for therapeutic intervention.. Clinical cancer research : an official journal of the American Association for Cancer Research(IF=10.2). 2008. PMID:18245520. DOI: 10.1158/1078-0432.CCR-07-2220.
- [40] Arnaud Descot;Thordur Oskarsson. The molecular composition of the metastatic niche.. Experimental cell research(IF=3.5). 2013. PMID:23707205. DOI: .
- [41] Yadi Wu;Binhua P Zhou. Inflammation: a driving force speeds cancer metastasis.. Cell cycle (Georgetown, Tex.)(IF=3.4). 2009. PMID:19770594. DOI: 10.4161/cc.8.20.9699.
- [42] Lei Zhang;Xichu Duan;Yanhua Zhao;Dejiu Zhang;Yuan Zhang. Implications of intratumoral microbiota in tumor metastasis: a special perspective of microorganisms in tumorigenesis and clinical therapeutics.. Frontiers in immunology(IF=5.9). 2025. PMID:39995663. DOI: 10.3389/fimmu.2025.1526589.
- [43] Christiana M Neophytou;Myrofora Panagi;Triantafyllos Stylianopoulos;Panagiotis Papageorgis. The Role of Tumor Microenvironment in Cancer Metastasis: Molecular Mechanisms and Therapeutic Opportunities.. Cancers(IF=4.4). 2021. PMID:33922795. DOI: 10.3390/cancers13092053.
- [44] Daniela Spano;Massimo Zollo. Tumor microenvironment: a main actor in the metastasis process.. Clinical & experimental metastasis(IF=3.2). 2012. PMID:22322279. DOI: 10.1007/s10585-012-9457-5.
- [45] Heath A Smith;Yibin Kang. The metastasis-promoting roles of tumor-associated immune cells.. Journal of molecular medicine (Berlin, Germany)(IF=4.2). 2013. PMID:23515621. DOI: 10.1007/s00109-013-1021-5.
- [46] Zhongnan Yin;Chunxiao Li;Junjie Wang;Lixiang Xue. Myeloid-derived suppressor cells: Roles in the tumor microenvironment and tumor radiotherapy.. International journal of cancer(IF=4.7). 2019. PMID:29992569. DOI: 10.1002/ijc.31744.
- [47] Yang Gao;Jeffrey M Rosen;Xiang H-F Zhang. The tumor-immune ecosystem in shaping metastasis.. American journal of physiology. Cell physiology(IF=4.7). 2023. PMID:36717100. DOI: 10.1152/ajpcell.00132.2022.
- [48] Thomas Bogenrieder;Meenhard Herlyn. Axis of evil: molecular mechanisms of cancer metastasis.. Oncogene(IF=7.3). 2003. PMID:14528277. DOI: 10.1038/sj.onc.1206757.
- [49] Emmanuel Viala;Jacques Pouysségur. Regulation of tumor cell motility by ERK mitogen-activated protein kinases.. Annals of the New York Academy of Sciences(IF=4.8). 2004. PMID:15659800. DOI: 10.1196/annals.1329.027.
- [50] Mateusz Kciuk;Adrianna Gielecińska;Adrianna Budzinska;Mariusz Mojzych;Renata Kontek. Metastasis and MAPK Pathways.. International journal of molecular sciences(IF=4.9). 2022. PMID:35409206. DOI: 10.3390/ijms23073847.
- [51] Abeer Alharbi;Yuxuan Zhang;John Parrington. Deciphering the Role of Ca2+ Signalling in Cancer Metastasis: From the Bench to the Bedside.. Cancers(IF=4.4). 2021. PMID:33430230. DOI: 10.3390/cancers13020179.
- [52] Frank J Lowery;Dihua Yu. Growth factor signaling in metastasis: current understanding and future opportunities.. Cancer metastasis reviews(IF=8.7). 2012. PMID:22706845. DOI: 10.1007/s10555-012-9380-x.
- [53] Mohammed Kaleem;Lubna Azmi;Naiyer Shahzad;Murtada Taha;Shiv Kumar;Md Ali Mujtaba;Abdulaziz Ali H Hazazi;Asaad Kayali. Epigenetic dynamics and molecular mechanisms in oncogenesis, tumor progression, and therapy resistance.. Naunyn-Schmiedeberg's archives of pharmacology(IF=3.1). 2025. PMID:40358685. DOI: 10.1007/s00210-025-04217-5.
- [54] Hui Ming;Bowen Li;Li Zhou;Ajay Goel;Canhua Huang. Long non-coding RNAs and cancer metastasis: Molecular basis and therapeutic implications.. Biochimica et biophysica acta. Reviews on cancer(IF=8.3). 2021. PMID:33548345. DOI: 10.1016/j.bbcan.2021.188519.
- [55] Maxime Janin;Veronica Davalos;Manel Esteller. Cancer metastasis under the magnifying glass of epigenetics and epitranscriptomics.. Cancer metastasis reviews(IF=8.7). 2023. PMID:37369946. DOI: 10.1007/s10555-023-10120-3.
- [56] Die Hu;Tianci Zhao;Chenxing Xu;Xinyi Pan;Zhengyu Zhou;Shengjie Wang. Epigenetic Modifiers in Cancer Metastasis.. Biomolecules(IF=4.8). 2024. PMID:39199304. DOI: 10.3390/biom14080916.
- [57] Rakesh Banerjee;Jim Smith;Michael R Eccles;Robert J Weeks;Aniruddha Chatterjee. Epigenetic basis and targeting of cancer metastasis.. Trends in cancer(IF=17.5). 2022. PMID:34952829. DOI: 10.1016/j.trecan.2021.11.008.
- [58] Antonio Mazzocca;Vinicio Carloni. The metastatic process: methodological advances and pharmacological challenges.. Current medicinal chemistry(IF=3.5). 2009. PMID:19442141. DOI: 10.2174/092986709788186192.
- [59] Xiaoli Shi;Xinyi Wang;Wentao Yao;Dongmin Shi;Xihuan Shao;Zhengqing Lu;Yue Chai;Jinhua Song;Weiwei Tang;Xuehao Wang. Mechanism insights and therapeutic intervention of tumor metastasis: latest developments and perspectives.. Signal transduction and targeted therapy(IF=52.7). 2024. PMID:39090094. DOI: 10.1038/s41392-024-01885-2.
- [60] Ana Hipólito;Filipa Martins;Cindy Mendes;Filipa Lopes-Coelho;Jacinta Serpa. Molecular and Metabolic Reprogramming: Pulling the Strings Toward Tumor Metastasis.. Frontiers in oncology(IF=3.3). 2021. PMID:34150624. DOI: 10.3389/fonc.2021.656851.
- [61] Adriana G Quiroz-Reyes;Jose F Islas;Paulina Delgado-Gonzalez;Hector Franco-Villarreal;Elsa N Garza-Treviño. Therapeutic Approaches for Metastases from Colorectal Cancer and Pancreatic Ductal Carcinoma.. Pharmaceutics(IF=5.5). 2021. PMID:33466892. DOI: 10.3390/pharmaceutics13010103.
- [62] Tania Ray;Terry Ryusaki;Partha S Ray. Therapeutically Targeting Cancers That Overexpress FOXC1: A Transcriptional Driver of Cell Plasticity, Partial EMT, and Cancer Metastasis.. Frontiers in oncology(IF=3.3). 2021. PMID:34540690. DOI: 10.3389/fonc.2021.721959.
- [63] Nicolo Riggi;Michel Aguet;Ivan Stamenkovic. Cancer Metastasis: A Reappraisal of Its Underlying Mechanisms and Their Relevance to Treatment.. Annual review of pathology(IF=34.5). 2018. PMID:29068753. DOI: 10.1146/annurev-pathol-020117-044127.
- [64] Li Yang;P Charles Lin. Mechanisms that drive inflammatory tumor microenvironment, tumor heterogeneity, and metastatic progression.. Seminars in cancer biology(IF=15.7). 2017. PMID:28782608. DOI: 10.1016/j.semcancer.2017.08.001.
- [65] Yongxing Li;Fengshuo Liu;Qingjin Cai;Lijun Deng;Qin Ouyang;Xiang H-F Zhang;Ji Zheng. Invasion and metastasis in cancer: molecular insights and therapeutic targets.. Signal transduction and targeted therapy(IF=52.7). 2025. PMID:39979279. DOI: 10.1038/s41392-025-02148-4.
- [66] Zhuo Li;Yibin Kang. Emerging therapeutic targets in metastatic progression: A focus on breast cancer.. Pharmacology & therapeutics(IF=12.5). 2016. PMID:27000769. DOI: 10.1016/j.pharmthera.2016.03.003.
- [67] David G DeNardo;Magnus Johansson;Lisa M Coussens. Immune cells as mediators of solid tumor metastasis.. Cancer metastasis reviews(IF=8.7). 2008. PMID:18066650. DOI: 10.1007/s10555-007-9100-0.
- [68] Karuna Ganesh;Joan Massagué. Targeting metastatic cancer.. Nature medicine(IF=50.0). 2021. PMID:33442008. DOI: 10.1038/s41591-020-01195-4.
- [69] L Lin;H Huang;W Liao;H Ma;J Liu;L Wang;N Huang;Y Liao;W Liao. MACC1 supports human gastric cancer growth under metabolic stress by enhancing the Warburg effect.. Oncogene(IF=7.3). 2015. PMID:25043301. DOI: 10.1038/onc.2014.204.
- [70] Amaia Lujambio;Manel Esteller. How epigenetics can explain human metastasis: a new role for microRNAs.. Cell cycle (Georgetown, Tex.)(IF=3.4). 2009. PMID:19177007. DOI: 10.4161/cc.8.3.7526.
- [71] Xiao-Yuan Mao;Qiu-Qi Li;Yuan-Feng Gao;Hong-Hao Zhou;Zhao-Qian Liu;Wei-Lin Jin. Gap junction as an intercellular glue: Emerging roles in cancer EMT and metastasis.. Cancer letters(IF=10.1). 2016. PMID:27490999. DOI: .
MaltSci Intelligent Research Services
Search for more papers on MaltSci.com
cancer metastasis mechanisms · tumor microenvironment · molecular pathways
© 2025 MaltSci
