Preprint
Review

This version is not peer-reviewed.

Mechanisms of Cancer Metastasis: Dormancy, Immune Evasion, Pre-Metastatic Niches, and Emerging Therapeutic Strategies

A peer-reviewed article of this preprint also exists.

Submitted:

29 November 2025

Posted:

03 December 2025

You are already at the latest version

Abstract

Metastatic relapse often reflects the survival of a small population of disseminated tumor cells (DTCs) that take up residence in distant organs and then shift into a dormant state. Rather than dividing, these cells sit quietly for long periods and rely on local niche signals to stay inactive and avoid therapy. Dormant cells are difficult to eliminate because the immune system cannot detect them, and treatments aimed at actively growing cells are ineffective. DTCs stop oncogenic signaling and start stress-response and cell-cycle arrest pathways. These pathways are often characterized by higher levels of cyclin-dependent kinase (CDK) inhibitors and greater p38 signaling than ERK signaling. LIFR–STAT3 signaling in the bone marrow supports quiescence in breast cancer cells, while inflammatory cytokines and Wnt/BMP antagonists in the lung microenvironment can trigger reactivation of cancer DTCs. Because these dormant DTCs are not cycling, standard cytotoxic agents rarely remove them. Current strategies are now testing immune-directed therapies. Recent single-cell and long-read sequencing efforts have started to reveal the transcriptional programs that mark DTCs, including stress-response and quiescence signatures that differ from the primary tumor. These insights are shaping therapies for interrupting dormancy and lowering the risk of late metastatic relapse.

Keywords: 
;  ;  ;  ;  ;  ;  ;  ;  ;  ;  

Introduction

Metastasis, which is the spread and growth of cancer cells in other organs, is the most dangerous part of cancer and causes most cancer deaths1. Many primary tumors do not respond favorably to surgery or radiation; however, metastasis is a systemic process in which tumor cells disseminate via blood or lymphatic vessels and subsequently adapt, survive, and proliferate in remote organs2. The dynamic alteration of tumor cell behavior and their manipulation of adjacent tissues and immune cells is crucial for effective metastatic dissemination. The distinct responses of primary and metastatic tumors to treatment underscore the unique biology and resistance associated with metastatic disease3. Modern sequencing technologies have changed how we think about metastasis. Genetic profiling of primary tumors, circulating tumor DNA, and circulating tumor cells has unveiled significant variation and evolutionary distinctions between primary and metastatic lesions4. These technologies allow for real-time monitoring of diseases, accelerate the search for biomarkers and treatment targets, and reveal how cancer cells spread by using host tissues to live and grow5.
Dormancy is a long-standing way for organisms to survive stress, and cancer cells seem to use this evolutionary program to wait out bad conditions6. When T cells enter a resting memory state, similar pathways are activated in immune cells. Tumor dormancy appears to hijack these conserved programs, allowing malignant cells to endure adverse conditions, such as immune pressure and toxic treatments, by entering a reversible growth arrest7. Dormancy may manifest locally before the proliferation of the primary tumor or systemically before the development of evident metastasis. Metastatic dormancy may occur before the detection of primary tumors. Recent data contradict the classical stepwise model, demonstrating that dissemination can occur very early, even from tumors smaller than 5 millimeters, followed by extended dormancy at remote locations6. Dormant cells can be single cells, small groups of cells, or tiny lesions that stay the same size because of balanced cell division and death8. The cancer cell’s own survival pathways, like autophagy and stress response systems, as well as physical factors in the environment, like the composition of the tissue scaffolding, the stiffness of the tissue, and the availability of growth factors, control dormancy8, 9. Metastasis is now recognized as a complex systemic process necessitating collaboration between tumor cells and host tissue, encompassing tissue remodeling, immune evasion, angiogenesis, metabolic adaptability, and intercellular cooperation among various tumor cell types2.
Finding hidden metastases early is a big problem when it comes to treating cancer. Evidence suggests that dissemination may transpire unexpectedly early in tumor development, potentially preceding clinical diagnosis. New blood tests, like the liquid biopsy platform, that look for circulating tumor cells, tumor DNA, or RNA-containing vesicles, show promise for catching minimal disease in real time1011. These tools are beginning to show how early metastatic colonies form and change when used with single-cell genetic analysis and spatial mapping techniques5. When scans show metastases, they often look different genetically from the original tumor. This is because of pressures on specific organs, differences in local tissue, and changes that happen during treatment3. This causes significant differences between metastases and even within the same metastases, making standard lab models less useful and treatment responses harder to predict. Modern metastasis treatment increasingly necessitates personalized strategies predicated on biomarkers that correspond to the evolving biology of disseminated cancer cells and their microenvironment11. There are two main goals for improving metastasis treatment: killing dormant cancer cells and stopping them from coming back8. Immune-based methods, such as reprogramming tumor-associated immune cells, activating natural killer cells, or making cancer cells more visible to the immune system, show promise for getting rid of dormant and early metastatic cells2, 12. Making markers that are specific to dormancy will be important for early intervention, customizing preventive therapy, and, in the end, lowering the risk of metastatic relapse8.

1. Metastasis-Inducing Genes and Mechanisms

One of the hardest things about cancer is that it can spread to other parts of the body. It happens when cancer cells can leave where they started, travel through the bloodstream, and settle in new places. Cells must change their shape and behavior, survive harsh conditions, and adapt to very different environments throughout the body. This journey isn’t easy. Scientists need to understand how all of this works so they can develop ways to stop cancer from spreading. Snail transcription factors are a group of proteins that help cancer cells make these changes. Snail1, Snail2 (also called Slug), and Snail3 are the three types. They all help initiate a process that converts epithelial cells (which usually adhere in organized tissues) into mesenchymal cells (which can move more freely). Each one does something a little different. This change is an important step that allows cancer cells to move and invade other cells.
A. SNAIL1: The Master Orchestrator of the Epithelial to Mesenchymal Transition (EMT) Snail1 is the leading player in starting EMT. It works by turning off genes that keep epithelial cells together, like E-cadherin, and changing the cell’s structure so that it can move and invade more easily13. Snail1 does this by binding to specific DNA sequences in the E-cadherin gene, preventing its transcription. Studies in mice show that Snail1 is essential at many stages of metastasis14. When cancer cells express active Snail1, they are much better at entering blood vessels and forming new tumors in the lungs. But Snail1 doesn’t just change the cancer cells. It also alters the surrounding tissue by turning normal support cells (fibroblasts) into cancer-supporting fibroblasts (cancer-associated fibroblasts, or CAFs). These cells have a very pro-invasive and inflammatory phenotype that helps the tumor grow and spread. These changed fibroblasts make prostaglandin E2 and enzymes that break down the scaffolding between cells. This makes it easier for cancer to move through tissues. This makes the environment stiffer, which activates some signaling pathways in the tumor cells and helps them stay hidden from the immune system15. Snail1 controls the spread of cancer by directly altering tumor cells and their environment.
B. SNAIL2 (SLUG): Stemness and Resistance to Treatment
Snail2 shares many features with Snail1 in terms of structure. It also stops epithelial adhesion molecules like E-cadherin, which start EMT programs that help cells move. Even though there is some overlap, Snail2 has functional traits that set it apart from other Snail family members16. One crucial difference is that it significantly affects the traits of cancer stem cells and their response to treatment. In breast cancer, elevated Snail2 levels are closely linked to recurrence and metastatic spread, highlighting its role in maintaining stem-like populations that can endure long-term and subsequently regenerate metastatic lesions17. Snail2 also helps tumors spread to other parts of the body by making tumor cells more likely to survive while they are circulating and making it easier for them to settle in new places18. This is true for melanoma, colorectal cancer, glioblastoma (GBM), and hepatocellular carcinoma (HCC). While Snail2 can alter stromal cell organization, its roles are less clear than those of Snail1. Snail3, on the other hand, is more involved in immune and developmental pathways and only plays a small role in the progression of metastasis19.
C. The TWIST Family: Specialization in EMT and Invasion
The Twist family of basic helix-loop-helix (bHLH) transcription factors includes Twist1 and Twist2. Both of these proteins are often inappropriately activated in cancer, and they play essential roles in the spread of cancer cells through distinct yet complementary mechanisms20.
D. TWIST1: Master EMT Inducer and Invasion Coordinator Twist1 is a key regulator of EMT. It stops E-cadherin and starts a wide range of transcriptional changes that make cells less sticky, change the structure of the cytoskeleton, and make cells move more easily20. Twist1 not only plays a key role in EMT but also activates PDGFRα-Src signaling, which drives invadopodia formation. These are actin-rich protrusions that bring together membrane-type 1 matrix metalloproteinase (MT1-MMP), MMP2, and MMP9 to create focused proteolysis at the leading edge. This way, Twist1 helps tumor cells break through basement membranes and move into the stroma around them21. The increased expression of Twist1 in breast cancer and osteosarcoma (OS) is linked to more aggressive metastasis and worse clinical outcomes. This shows that it is an important prognostic marker for metastasis in many types of tumors22.
E. TWIST2: Improving survival and changing the immune system
Twist2, while less well known than Twist1, plays essential roles in strengthening EMT programs and helping tumor cells survive. It boosts the levels of anti-apoptotic regulators like Bcl-2 and survivin, which makes treatment less effective and leads to a disease that keeps coming back23. Twist2 also affects critical signaling pathways like NF-κB and STAT3, which help create conditions in the tumor microenvironment (TME) that suppress the immune system and make it harder for the immune system to find and kill the tumor24.
F. Specificity Protein 1 (SP1): Master Transcriptional Coordinator Specificity Protein 1 (SP1) is a zinc-finger transcription factor with a wide range of pro-metastatic effects. It promotes EMT by activating major EMT regulators such as Snail, Twist, and ZEB1, and it increases the expression of matrix-modifying enzymes, including MMP-2, MMP-9, and MT1-MMP, that support extracellular matrix degradation and tissue invasion25. In addition to EMT regulation, Sp1 stimulates angiogenesis by directly inducing VEGF and PDGF, thereby supporting the vascular expansion required for tumor growth and metastatic spread26. Sp1 enhances metastatic fitness by increasing the levels of survival factors such as Bcl-2 and survivin, making cells more resistant to treatments that kill cancer cells27. Oncogenic signaling pathways, such as EGFR/ERK and PI3K/AKT, stabilize Sp1 and create feedback loops that enhance its activity28. This enhances its transcriptional output. Increased Sp1 expression is consistently associated with poor outcomes in multiple malignancies, such as breast, lung, colorectal, pancreatic, and liver cancers, highlighting its critical role in cancer progression29.

2. Epigenetic Regulators of Metastasis

A wide variety of epigenetic regulators directly promote metastasis. EZH2, the catalytic subunit of PRC2, is one of the most important of these. EZH2 represses tumor-suppressive genes, such as E-cadherin, by depositing H3K27me3. It also strengthens EMT by supporting transcription factors like Snail and Slug30, 31. EZH2 overexpression is significantly associated with an unfavorable prognosis in various carcinomas32. EZH2 diminishes epithelial integrity by silencing adhesion genes, facilitating invasion, and inducing fibroblast-to-myofibroblast conversion and wound-healing stromal remodeling that further enhances metastasis33. Various non-epigenetic oncogenic drivers also facilitate metastatic progression. PTTG1 (securin) encourages genomic instability and increases invasive behavior34. BIRC5 (survivin) helps circulating tumor cells stay alive by stopping apoptosis35. YBX1 controls EMT, stemness, and treatment resistance36. Transcription factors like E2F1 and MYB, which control the cell cycle, also turn on gene programs that are involved in invasion37, 38. These factors facilitate invasion, survival, and microenvironmental remodeling during metastasis. In the end, epigenetic reprogramming is crucial for metastatic competence because it gives cells the flexibility to invade, spread, remain dormant, and form new tumors.
DNA Methylation: Abnormal methylation alters genes that control tumor growth. DNMT1, DNMT3A, and DNMT3B inhibit adhesion molecules like CDH1 and enhance the expression of EMT transcription factors such as Snail, Twist, and ZEB39. Global hypomethylation at repetitive sequences and enhancers also activates oncogenic and inflammatory pathways that promote dissemination40.
Histone Methylation: EZH2-driven H3K27me3 inhibits tumor suppressors, lineage regulators, and immune-related genes30, 41. Other methyltransferases, like G9a (EHMT2; H3K9) and SUV39H1, keep epithelial identity from being expressed and help keep mesenchymal states42. The absence of demethylases like KDM6A (UTX) or KDM6B (JMJD3) enhances the stability of epithelial-mesenchymal transition (EMT) programs and facilitates metastasis43.
Histone acetylation and deacetylation: Histone acetyltransferases (p300/CBP) activate EMT drivers44 and, when metastatic disease is present, they stop epithelial programs while boosting angiogenesis and immune evasion45. HDAC inhibitors can reverse EMT characteristics and enhance responses to immunotherapy46.
Chromatin Remodeling: Changes in SWI/SNF (BAF) complexes affect metastasis, depending on the situation. The absence of ARID1A or SMARCA4 enhances dissemination in various cancers, whereas different BAF configurations encourage invasion and therapeutic resistance47.
Noncoding RNAs and Interactions: Long noncoding RNAs and microRNAs regulate chromatin-modifying enzymes. HOTAIR, for instance, brings PRC2 to shut down epithelial genes and speeds up EMT and distant metastasis in breast cancer48. The reversibility of epigenetic modifications presents therapeutic prospects in clinical settings. Targeting epigenetic plasticity thus constitutes a promising strategy to halt or treat metastatic progression. Human cancer studies have linked each of the genes above to cancer spread, usually through the mechanisms described. For example, Snail1 and Twist1 have been shown to directly cause invasion through EMT programs. IL-6 and IL-8 are well-known cytokines that promote metastatic phenotypes in many cell types, and MMPs degrade the extracellular matrix, enabling cells to spread.
Noncoding RNAs and Crosstalk: Long noncoding RNAs and microRNAs coordinate chromatin-modifying enzymes. HOTAIR, for example, recruits PRC2 to silence epithelial genes and promotes EMT and distant metastasis in breast cancer. Clinically, the reversibility of epigenetic alterations offers therapeutic opportunities49, 50. Targeting epigenetic plasticity, therefore, represents a promising approach to interrupt or treat metastatic progression.
Table 1. Key genes and their primary mechanisms driving tumor metastasis.
Table 1. Key genes and their primary mechanisms driving tumor metastasis.
Gene name Function Description REF
Snail EMT, CAF activation, Prostaglandin E2 (PGE2) Induces EMT and invasion in carcinoma cells (Represses E-cadherin). Required for CAF activation; CAFs secrete PGE₂ and cytokines to drive tumor invasion (Regulates mesenchymal differentiation, wound healing) 51
TWIST1 EMT, PDGFRα→Src, invadopodia TWIST1 EMT TF: invadopodia formation (via PDGFRα/Src) Drives EMT, motility, invadopodia-mediated ECM degradation (Not directly studied). Upregulated in CAFs of many tumors: Twist1 promotes invasion and tumor growth. 21
SP1 Pan-cancer TF, survival/invasion, WNT/β-catenin Pan-cancer TF; induces WNT signaling, survival, and invasion. Master regulator of metastasis genes; enhances WNT/β-catenin signaling in tumor cells. Drives expression of angiogenic factors (e.g., VEGF); WNT signals from stroma to endothelium. 52, 53
IL-6 STAT3/EMT; CAF source; angiogenesis Pro-inflammatory cytokine; activates JAK/STAT3, EMT receptor-expressing carcinoma cells undergo STAT3-dependent EMT and proliferation. Promotes angiogenesis and leukocyte recruitment in tumor vessels. Secreted by CAFs (and tumor cells); drives EMT/migration of cancer cells. IL-6 can recruit and modulate MSCs (MSC chemotaxis, differentiation) 54, 55
CXCL8 (IL-8) Angiogenesis, EMT/invasion, CXCR1/2 Promotes angiogenesis, EMT, and invasion. Tumor-derived IL-8 induces autocrine EMT/invasion and survival. Potent angiogenic factor; stimulates endothelial proliferation and vessel permeability. CAFs secrete IL-8 to boost tumor angiogenesis and invasion. MSCs respond to IL-8 (via CXCR1/2), promotes MSC migration and possibly MSC-to-CAF transition. 56, 57
CXCL1 Neutrophil recruitment, angiogenesis CXCL1 Chemokine (ELR+); recruits neutrophils, fosters angiogenesis. Tumor-secreted CXCL1 creates a pro-inflammatory niche for invasion (by analogy to IL-8). Angiogenic; contributes to neovascularization (via CXCR2). Expressed by CAFs and TAMs; enhances tumor cell motility and chemoresistance (paracrine). May attract MSCs to the tumor; role is less defined than IL-8 58, 59
CXCR4 CXCL12 homing, organotropism CXCR4 Chemokine receptor; guides cells to CXCL12-rich organs. Binds CXCL12 to direct cancer cell homing/migration to metastatic sites (lung, liver, bone). Endothelial cells produce CXCL12; CXCR4+ tumor cells adhere to the vasculature and extravasate. CXCR4 is expressed on fibroblasts/CAFs; CXCL12 from stroma promotes tumor-CAF interactions. Highly expressed on MSCs; mediates MSC homing and survival. 60, 61
MMP9 ECM degradation, growth-factor activation, angiogenesis MMP9 Secreted matrix metalloprotease; cleaves ECM, activates growth factors. Tumor cells secrete MMP9 to breach the basement membrane (promoting intravasation). Degrades endothelial basement membranes to enable angiogenesis and metastasis. CAFs/myofibroblasts produce MMP9 to remodel the stroma and release pro-metastatic signals. MSCs secrete MMP9 to facilitate migration; MSC-derived MMPs shape the metastatic niche. 62, 63
MMP1 Interstitial collagenase, invasion/angiogenesis MMP1 Interstitial collagenase; degrades type-I/III collagen. Tumor-derived MMP1 promotes invasion through dense stroma, enabling new vessel growth by remodeling perivascular ECM. CAFs produce MMP1 to stiffen or remodel the matrix, enabling tumor spreading. MSCs may also express MMP1 in differentiation contexts. 64, 65
EZH2 H3K27me3 silencing, EMT, stromal remodeling EZH2 Histone methyltransferase; epigenetic silencer of adhesion genes. Silences E-cadherin/epithelial genes, activating EMT and invasion (May promote EndMT by methylating endothelial promoters). Drives fibroblast-to-myofibroblast transition; promotes fibrotic stroma. Regulates MSC proliferation/differentiation (Wound healing analogies). 66, 67
FOXM1 EMT, MMPs, angiogenesis FOXM1 Forkhead TF; drives cell cycle, EMT (upregulates Snail/MMPs). In tumor cells, FOXM1 induces MMP2/9 and EMT factors, enhancing invasion. Promotes angiogenesis via VEGF expression; also implicated in EndMT in fibrosis. Shown to regulate CAF proliferation and extracellular proteases (in some tumors). May influence MSCs’ proliferative and migratory potential FOSL1 (FRA1) AP-1 subunit; EMT and invasion activator. Upregulates genes involved in motility (e.g., MMPs); promotes a mesenchymal phenotype. Stimulates VEGF and inflammatory cytokines, aiding vessel formation. In stromal cells, it supports the production of pro-tumorigenic ECM factors. In MSCs, differentiation may tilt toward a CAF-like state. 68, 69
E2F1 Cell cycle invasion/angiogenesis programs E2F1 Cell-cycle TF; pro-metastatic when overexpressed. Aside from proliferation, E2F1 can induce MMPs and EMT-associated genes. Drives expression of angiogenic factors (FGF, VEGF); can act in the endothelium. Linked to fibroblast proliferation; may contribute to desmoplasia. Activates proliferation of MSCs and endothelial precursors. 70
MYB stemness/invasion; angiogenic transcription MYB Transcription factor can promote stemness and invasion. Activates target genes (including MMPs, EMT factors) in carcinomas. Regulates angiogenic gene expression (e.g., VEGFR). Influences fibroblast proliferation; MYB is expressed in some CAF subsets. Helps maintain MSC self-renewal; influences differentiation pathways. 71
PTTG1 (Securin) Genomic instability, EMT, invasion PTTG1 (Securin) Promotes genetic instability and EMT. Overexpressed PTTG1 drives EMT and cell motility in cancer cells. May enhance secretion of angiogenic factors (through p53 inhibition). In fibroblasts, PTTG1 can promote proliferation and matrix production. In MSCs, PTTG1 supports proliferation, possibly aiding their tumorigenic roles. 72, 73
YBX1 EMT, stress survival, drug resistance YBX1 RNA/DNA-binding protein; induces EMT and stress survival. Activates EMT-related mRNAs (Snail, Twist) and drug resistance pathways in tumors. Regulates VEGF expression under hypoxia, promoting angiogenesis. Contributes to fibroblast activation by stabilizing cytokine mRNAs. Modulates MSC plasticity and response to microenvironmental stress. 74, 75
BIRC5 (Survivin) Anoikis resistance, survival of CTCs/ endothelium BIRC5 (Survivin) Inhibitor of apoptosis; cell division regulator. Upregulated in metastatic tumors to allow anoikis resistance and survival in circulation. Supports the survival of proliferating endothelium in tumor vessels. Protects CAFs/myofibroblasts from apoptosis, sustaining pro-metastatic stroma. Ensures MSC survival in harsh metastatic niches. 76, 77
ZEB2 EMT TF; metastasis and stromal/EndMT links ZEB2 EMT transcription factor; represses epithelial genes. Drives EMT and mesenchymal phenotype in carcinoma cells (analogous to Snail/Zeb1) (Possible role in EndMT/transdifferentiation of endothelium). Induces fibroblast-like program in epithelial and endothelial cells. In MSCs, ZEB2 may regulate multilineage differentiation toward mesenchyme. 78, 79
Each gene above is implicated by human cancer studies in promoting metastasis, often through the described mechanisms. For instance, Snail1 and Twist1 have been directly shown to stimulate invasion via EMT programs, IL-6 and IL-8 are well-known cytokines that activate metastatic phenotypes in multiple cell types, and MMPs remodel the extracellular matrix to permit dissemination.

3. Extracellular Proteases and Matrix Modifiers in Metastatic Progression

Recent accumulating evidence consistently identifies the extracellular matrix (ECM) as a pivotal factor influencing cancer risk and progression. For instance, women with breast tissue that is rich in collagen have a fourfold higher risk of getting breast cancer80. In established tumors, the ECM serves as both a structural framework and a source of biochemical signals. Metastatic cells release enzymes that degrade the ECM, such as extracellular proteases and matrix-modifying enzymes. These enzymes dismantle the matrix structure, liberate growth factors previously sequestered within the ECM, and produce biologically active fragments that facilitate invasion, angiogenesis, inflammation, and the establishment of pre-metastatic niches. Cross-species xenograft studies indicate that extracellular matrix (ECM) remodeling occurs through unique yet synergistic contributions from malignant and stromal compartments. Tumor cells primarily synthesize matrix-modifying and matrix-degrading enzymes, while stromal fibroblasts and other mesenchymal cells predominantly provide ECM glycoproteins; both cell types are involved in the production of fibrillar collagens81. Collagen I, specifically, is consistently elevated in primary tumors of the breast, liver, lung, and esophagus, and is similarly heightened in metastatic ovarian cancer82-84. Hyaluronic acid (HA) builds up in breast, prostate, bladder, and colon cancers and is linked to metastasis and a bad outcome85, 86. Elevated epithelial fibronectin in breast cancer indicates diminished survival. Laminin-111 is reduced in tumors compared to normal tissues87, whereas laminin-332 is elevated in various cancers88, including breast cancer, and is associated with a poorer prognosis. In advanced omental metastases of ovarian cancer, there are more glycoproteins (like fibrinogen and fibronectin), proteoglycans, secreted factors, and ECM-associated proteins than in smaller lesions89. Proteomic analyses have identified tenascin-C as a prognostic marker in lung cancer and SERPINB1 as a factor influencing brain tropism in breast cancer metastasis90. RGD-modified HA improves cooperative chemotherapy resistance in GBM91, PEG hydrogels with MMP-degradable linkers enable GBM cells to develop actin-rich protrusions, while non-degradable matrices confine cells to a rounded morphology91.
A. Functions of the Matrix Metalloproteinase Family
Matrix metalloproteinases (MMPs) are the most well-known extracellular proteases in cancer. MMP-9, a highly active gelatinase, is abundant in aggressive tumors. It releases growth factors like VEGF and TGF-β that are bound to the matrix, thereby strengthening signals that drive invasion and angiogenesis. MMP-1, an interstitial collagenase, cleaves fibrillar collagens, making it easier for stromal cells to enter tissues and penetrate them92. MMP-9 facilitates various stages of metastatic progression, encompassing EMT induction, enhanced cell migration, angiogenic activation, and the sustenance of tumor-associated inflammatory responses93. MMP-9 remodels collagen structure and promotes endothelial sprouting at invasive fronts. This makes neovascularization easier, which is necessary for tumor growth and spread94. MMP-9 from tumors facilitates intravasation, while MMPs from CAFs alter the stroma, making it stiffer to help the tumor keep invading95.
B. Additional Protease Systems
High uPA pathway activity is consistently associated with poorer prognosis in breast and colorectal cancers, underscoring its clinical significance96. This system performs necessary supplementary functions to MMP systems by degrading fibrillar collagens and components of the basement membrane. Cathepsins, particularly cathepsins B, L, and S, enhance matrix-degrading capabilities and are instrumental in compromising vascular barriers, thereby facilitating intravasation and aiding metastatic dissemination97.
C. Enzymes that change the matrix and control biomechanics
Tumor cells release specialized matrix-modifying enzymes that alter the biomechanical properties of the ECM, thereby accelerating cancer spread. The lysyl oxidase (LOX) family of enzymes links collagen fibers together, which makes the matrix stiffer. This rigidity triggers mechanosensitive signaling pathways that include integrins, FAK, and RhoA, which help cells move. In breast cancer research, hypoxic conditions stimulate LOX, which remodels collagen and creates a pre-metastatic niche in the lung98. LOXL2, a related enzyme, stabilizes EMT transcription factors like Snail by chemically changing them after they are made. This strengthens mesenchymal traits and makes cells more likely to spread to other parts of the body99. Heparanase alters the extracellular matrix (ECM) by cleaving heparan sulfate proteoglycans, which releases angiogenic and pro-invasive factors like FGF and VEGF and makes it easier for tumor cells to move around100.

4. Mechanisms of Immune Evasion in Metastatic Cancer

Immune evasion during metastasis occurs via multiple overlapping mechanisms, including suppressive immune cells, checkpoint signals, and physical barriers within the tissue. Monocytic myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) inhibit the immune system by secreting molecules such as TGF-β, reactive oxygen species, and arginase, which diminish the cytotoxic capacity of T cells and natural killer (NK) cells. TAMs frequently transition to an alternatively activated M2-like phenotype that promotes angiogenesis and facilitates tumor evasion of immune surveillance101. Platelets help hide tumor cells from the immune system by coating them as they move through the blood. Platelets release thromboxane A2, which can stop T-cells from becoming active through the ARHGEF1 signaling pathway. Targeting platelets with antiplatelet drugs significantly impairs the efficiency of metastatic cells in establishing new tumors, indicating the potential efficacy of this strategy in patients102. Within established metastases, T-cell dysfunction is exacerbated by various immune checkpoint pathways. In metastases, especially in protected areas like the brain, tumor-infiltrating T cells often have a lot of inhibitory receptors like PD-1, LAG-3, and TIGIT103. Stromal barriers that are very thick in the tumor microenvironment also prevent immune cells from reaching cancer cells. A thick extracellular matrix and the presence of cancer-associated fibroblasts (CAFs) can physically prevent effector immune cells from reaching tumor sites. For instance, in pancreatic cancer, a dense stroma rich in hyaluronan impedes blood flow and prevents T cells from entering the tumor core. Research in the lab has shown that using enzymes to break down these matrix components can make it easier for drugs to enter cells and for T cells to enter the stroma. This is evidence that strategies to normalize the stroma could be helpful104.

5. Cytokine and Chemokine Networks in Metastatic Dissemination

Cytokines and chemokines are the main things that control the spread of cancer cells to other parts of the body. They create networks of communication that guide cancer cells, establish pre-metastatic niches, and coordinate complex interactions between cancer cells and host tissues.
A. Chemokine-Mediated Organ Tropism
The CXCL12-CXCR4 axis is the best-known chemokine pathway that controls how cancer cells migrate to other organs. It directs breast and prostate cancer cells to CXCL12-rich tissues, including the lung, liver, and bone marrow, where elevated chemokine concentrations establish conducive environments for incoming tumor cells105. The CCR7-CCL21 chemokine axis facilitates lymphatic dissemination, as CCL21 is continuously expressed by lymphatic endothelial cells and lymph node stromal cells, directing CCR7-expressing tumor cells towards lymphatic vessels and draining lymph nodes106. The CCR9-CCL25 axis also causes metastasis in the gut by directing tumor cells to intestinal sites that are rich in CCL25107.
B. Pro-Metastatic Cytokine Signaling
Cytokine networks facilitate metastasis via multiple coordinated pathways. Interleukin-6 (IL-6) activates JAK/STAT3 signaling, which promotes epithelial-mesenchymal transition (EMT) and cancer stem cell properties, as demonstrated in gastric cancer research examining the interaction between cancer-associated fibroblasts and tumor cells108. TGF-β has different effects depending on the situation. In early cancer, it stops tumors from growing, but in advanced cancer, it helps EMT, immune evasion, and metastasis109, 110. TNF-α enhances invasion by activating NF-κB, which elevates the expression of adhesion molecules and matrix metalloproteinases that facilitate cellular penetration and tissue remodeling. The collaboration of IL-1β and CXCL8/IL-8 activity attracts neutrophils, stimulates tumor angiogenesis, and sustains epithelial-mesenchymal transition (EMT) at the peripheries of invading tumors, thereby fostering inflammatory environments that facilitate tumor progression111.
C. Cytokine Networks in the Pre-Metastatic Niche
Certain cytokine combinations, such as VEGF-A, TNF-α, and TGF-β, act together to increase levels of S100A8/A9 and fibronectin in the lung’s supporting tissue during the formation of pre-metastatic niches. This methodically brings in myeloid cells to set up conditions that make it easier for tumor cells to settle later112. In patients, elevated levels of chemokines and cytokines are significantly associated with adverse outcomes, preferences for particular metastatic sites, and treatment resistance. Because of these results, clinical trials are now testing targeted therapies like CXCR4 inhibitors (plerixafor), CCR5 inhibitors (maraviroc, NCT01736813), IL-6 receptor antagonists (tocilizumab, NCT03999749), and TGF-β antagonists for the treatment of metastatic disease113.

6. Pre-Metastatic Niche Formation and Organ-Specific Colonization

A. Niche Preparation Orchestrated by the Primary Tumor
Primary tumors send signals that prepare other organs for cancer cells to move in long before they actually do. Significant research has shown that bone marrow cells expressing VEGFR1 travel to these distant sites in response to molecules released by the original tumor. This creates a support structure that makes it easier for metastases to grow later114. LOX is essential for this preparation because it crosslinks collagen molecules and attracts myeloid cells that remodel tissue structure in target organs like the lungs. It is made when tumors have low oxygen levels. VEGF and other chemokine signals, on the other hand, attract immune-suppressing cells that produce the environment better for metastatic cells115 to come in.
B. Communication Between Cells Through Exosomes
Exosomes released by primary tumors serve as communication vehicles that instruct resident fibroblasts and endothelial cells in remote organs on their behavior. These small vesicles also alter the function of circulating neutrophils and monocytes, turning them into cells that either help build new blood vessels or suppress immune responses. This makes it easier for metastatic cells to settle in later116.
C. Organ-Specific Niche Architecture and Upkeep
Pre-metastatic niches (PMNs) form in a way that is specific to the biology of each target organ. In the lung, early PMNs exhibit significant neutrophil clusters and infiltration by inflammatory macrophages. In bone, homing mediated by CXCR4-CXCL12 and signals that activate osteoclasts are very important. In the brain, PMN formation relies on specific adhesion interactions with endothelial cells and on alterations in the structure of the blood-brain barrier. Once these sites are ready, resident stromal cells help sustain metastatic growth through organ-specific mechanisms. In the lung, for example, perivascular fibroblasts produce tenascin-C, which attracts and activates macrophages. This strengthens a microenvironment that helps cancer cells spread117.

7. Tumor Dormancy

Tumor dormancy is a complex mechanism by which cancer cells can survive. They stay quiet, keep their metabolic activity low, and respond to signals from their surroundings. These changes allow tumor cells to survive for a long time, avoid the effects of treatment, and retain the ability to resume growth. Dormancy is not a passive state; it is sustained by coordinated gene expression, metabolism, epigenetic regulation, and signals from the local tissue environment that determine whether a cell remains quiescent or resumes division. Controlling dormancy depends a lot on the conditions in the area. Cells go into dormancy when there isn’t enough oxygen, nutrients, or blood vessels. Cells that try to divide in these harsh conditions usually die. Hypoxic stress, on the other hand, leads to aggressive traits and resistance to treatment118, 119. In the RIP1-Tag2 pancreatic islet tumor model, the removal of VEGF leads to highly hypoxic, slowly expanding tumors reliant on HIF1α for survival, while the inhibition of angiogenesis can enhance invasive characteristics12 120, 121. Cells that don’t get enough oxygen also resist treatment and can start tumors again, as shown by lineage tracing studies after radiation122. Low oxygen levels trigger HIF-driven glycolysis, but when cells lack both oxygen and glucose for a long time, they enter a dormant state in which their metabolism slows down123. In conditions of extreme oxygen deprivation, the removal of methyl marks from the HIGD1A gene promoter induces this protein, which facilitates survival by reducing mitochondrial respiration, activating AMPK, decreasing reactive oxygen species, and maintaining cellular viability124. Extracellular matrix (ECM) and integrin signaling also play a role in dormancy. The balance between ERK and p38 MAPK activity is a critical control point. When p38 signaling is stronger than ERK signaling following uPAR–α5β1 interactions, HEp3 carcinoma cells enter a dormant state125. In breast cancer, blocking β1-integrin causes dormancy126, and the amount of fibronectin deposited and the amount of tension in the cytoskeleton decide whether D2.0R cells stay dormant or D2A1 cells keep dividing in 3D culture127. Thrombospondin-1 released by endothelial cells128 is another signal from the surrounding tissue that keeps cells dormant. The lack of CXCR4 expression, observed in dormant lung metastases, is another signal. Changes in genes and epigenetics within the cell also help keep it dormant129. NR2F1 induces dormancy by activating SOX9, p27, and CDK inhibitor pathways130. In models derived from patient tumors, NR2F1 collaborates with HIF1α to maintain dormant cancer cells131. Dormant cells usually have fewer MHC molecules and antigen-processing parts, which makes it harder for T cells and NK cells to find them. At the same time, they turn on stress-response systems that help them live longer. In melanoma models, CD8+ T cells inhibit metastatic expansion, and the removal of immune cells leads to rapid tumor growth132. Dormancy is a major obstacle for doctors seeking cures. Spread cancer cells can stay dormant for decades in a quiet, treatment-resistant state. This is why breast, prostate, and other cancers sometimes come back years later133. Many molecular pathways control the switch between dormancy and reactivation. Proteins that change chromatin, like KDM5B, EZH2, KDM6A/KDM6B, and TET2, decide if chromatin is open or closed. Transcription factors such as c-JUN, FOXM1, NR2F1, NANOG, and SOX2, on the other hand, control transitions between the resting and dividing states134. The p38-NR2F1-p53 signaling pathway keeps cells dormant, while the ERK activation, uPAR-FAK/PI3K, and other pathways that promote growth push cells out of dormancy. Other ways to control things include protein degradation systems (FBXW7, SKP2), noncoding RNAs (NR2F1-AS1, ELEANOR, miRNAs), and signals from nearby tissues, such as WNT modulators, IL-6/LIFR–STAT3, and STING signaling135. There are three main types of treatment strategies. Reactivation strategies aim to induce dormant cells to re-enter the cell cycle, rendering them vulnerable to cytotoxic agents targeting proliferating cells. However, incomplete eradication of all cells may expedite tumor progression, as observed following the TGFβ/p38 in head and neck cancer. Dormancy-maintenance strategies aim to prolong cellular dormancy through the utilization of retinoids that activate NR2F1 and TGFβ2 signaling, STING activators, glutaminase inhibitors, anti-SFRP1 therapies, and signals derived from the extracellular matrix, such as type III collagen or factors from senescent tissue136-139. Eradication strategies focus on vulnerabilities specific to dormant cells, including YAP/TEAD activity, ROCK signaling, fibronectin-integrin interactions, and particular metabolic deficiencies140. Improvements in computational oncology are making it easier for us to find dormant disease. Combined models that use tissue microscopy, patient information, and blood tests such as ctDNA methylation, mutation profiles, and DNA fragment patterns can detect very small amounts of residual disease and predict relapse early141,142. Long-read sequencing and analysis of extrachromosomal DNA are expected to reveal superior markers at the isoform and structural levels that signify persistence and recurrence.

8. Tumor Metastasis Mechanisms by Tumor Type

Metastasis is when cancer cells, the surrounding tissue, and the immune system all work together in complicated ways. This is what kills most people with cancer. This review integrates organ- and tissue-specific metastatic patterns with their underlying biological mechanisms. These include how cancer cells change their behavior, how they alter nearby tissues and the extracellular matrix, how tumors evade immune responses, and how they prepare to spread to other parts of the body. This review talks about the most common tumors. The conclusion emphasizes clinical applications, including biomarkers that indicate the potential for cancer metastasis to other organs and treatment strategies that target the vicinity of cancer cells.

8.1. Osteosarcoma (OS)

Early metastasis, particularly to the lungs, is the principal factor influencing prognosis (Managing the immune microenvironment of OS and treatment). About 15-20% of patients have metastases at the time of diagnosis. Of those who are first diagnosed with localized disease, 30-40% will eventually have a relapse, most often with pulmonary disease. About 80-90% of distant metastases affect the lungs, with secondary bone involvement at approximately 8-11%, and, infrequently, visceral or CNS locations143-145 146. The lung is still the most common place for OS to come back and the most common cause of death from OS (The metastatic patterns of OS). At the genomic level, OS exhibits significant chromosomal instability and persistent disruption of established tumor-suppressor pathways147. The most common type of loss-of-function in TP53 and RB1 is when CDKN2A is deleted, and MYC/MDM2 is amplified148. These changes happen as part of complicated structural changes that help cancer cells adapt and spread. The Rab22a–NeoF1 fusion facilitates lung colonization by activating RhoA, remodeling the cytoskeleton, and forming invadopodia. Importantly, exosomes containing Rab22a-NeoF1 prepare pre-metastatic lung niches149. The histone demethylase KDM6B increases glycolytic flux by upregulating LDHA, which supports metastatic growth150. In the same way, the m6A RNA demethylases FTO and ALKBH5 control post-transcriptional programs that strengthen metastatic traits, such as making cells more invasive and better able to withstand stress151. Metastatic OS cells frequently demonstrate significant aerobic glycolysis (the Warburg effect), which promotes anabolic metabolism, supports survival in hypoxic environments, and leads to the acidification of the surrounding microenvironment152. The TME is therefore a central determinant of OS progression and metastatic competence153. TAMs, especially M2-polarized subsets, are associated with a poor prognosis and increased lung colonization because they release pro-angiogenic and immunosuppressive factors. Matrix metalloproteinases, especially MMP-9, help the ECM change shape and release growth factors154. Platelets shield circulating tumor cells from shear stress and immune surveillance, promoting vascular adhesion and metastatic seeding155. The pulmonary niche signals that help cells survive, and lung-derived mediators activate MAPK signaling and increase the levels of anti-apoptotic molecules like MCL1, which supports micrometastatic growth156. Metastatic competence reflects the integration of various oncogenic pathways. PI3K/AKT improves survival and metabolic adaptation; MAPK/ERK promotes proliferation and invasion; WNT/β-catenin controls stemness and motility; and Rho/Rac GTPases manage cytoskeletal remodeling157. Chemokine networks, especially the CXCL12/CXCR4/CCR7 axis, help tumor cells find their way to the right place. Integrins such as αVβ3 and αVβ5 help cells adhere firmly to the endothelium and the extracellular matrix. Blocking these integrins with drugs reduces the spread of cancer to the lungs in preclinical models158 159. Experimental orthotopic and intravenous models utilizing highly metastatic cell lines (e.g., K7M2, 143B) and genetically engineered murine models exhibiting conditional p53/Rb loss in osteoblasts have facilitated an in-depth analysis of the mechanisms underlying lung metastasis160 161. Clinical genomic datasets validate these findings, underscoring the significance of TP53/RB1 disruption and pulmonary relapse as primary contributors to treatment failure148. The immune microenvironment (IME) of OS, which includes T lymphocytes, natural killer (NK) cells, tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs), plays two roles: it helps the body fight cancer and helps it evade immune attack. Standard immunotherapies, including vaccines, cytokine therapy, and immune checkpoint blockade (ICB), have demonstrated limited effectiveness in OS. Emerging nano-immunotherapy strategies, on the other hand, aim to reverse the immunosuppressive IME, induce immunogenic cell death (ICD), and enable targeted delivery to improve treatment outcomes. Genomic changes, such as losing TP53 or RB1 and amplifying MYC or MDM2, work with changes in metabolism and epigenetics, such as KDM6B-mediated chromatin remodeling and a shift toward glycolytic metabolism, to make OS more likely to spread. These changes within the tumor interact with signals from the tumor’s microenvironment, which are generated by tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and activated platelets. Together, they help osteosarcoma cells spread and stay alive in other parts of the body. Pro-migratory signaling pathways (PI3K/AKT, MAPK, WNT/β-catenin, Rho/Rac) and chemokine–integrin networks (CXCL12/CXCR4/CCR7) work together to drive these processes. Rational combination strategies, including ICB with TAM/MDSC modulation, inhibition of chemokine or integrin axes, and nano-immunotherapy, represent promising therapeutic pathways. Future initiatives should emphasize biomarker-driven clinical translation, sophisticated delivery mechanisms, and single-cell/spatial analyses to address IME heterogeneity and surmount therapeutic resistance in OS. AXL is a key regulator of OS progression and is strongly associated with a bad prognosis162. It is a receptor tyrosine kinase (RTK). In murine models, AXL inhibition markedly diminished the pulmonary metastases from MG63.2 cells163. Clinical trials of multi-target tyrosine kinase inhibitors (TKIs) have demonstrated that AXL inhibition can elicit partial remission in some patients164. While these results are encouraging, no clinical trial has yet examined the impact of selective AXL inhibition alone on outcomes in OS patients. Preclinical studies further demonstrate that AXL knockdown inhibits OS cell proliferation and induces apoptosis165. AXL overexpression is observed in rapidly spreading, ubiquitously distributed OS cell lines, and blocking it slows tumor cell growth, invasion, and cell spread162. Fibroblast growth factor receptor (FGFR) is another RTK that is often overexpressed in OS. FGFRs control many bodily functions, such as the development of the nervous system, the formation of organs, and tissue healing166. FGFR1 is expressed in about 74% of OS samples, making it a very promising target for treatment167.
Figure 1. Key molecular drivers and microenvironmental determinants of OS metastasis in the lung. Molecular pathways in OS and lung microenvironmental elements that facilitate metastatic colonization. A combination of tumor-intrinsic signaling pathways, chemokine-integrin axes, and genomic changes drives OS metastasis. These changes make it easier for cancer cells to invade, intravasate, and colonize distant organs, mostly in the lungs. (Left) Signaling molecules that come from tumors, like WNT/β-catenin, MAPK, PI3K/AKT, RAB22A-NEOF fusions, and RhoA, make cells more mobile, help them survive, and make them better at spreading to other parts of the body. Chemokine networks, such as CXCL12/CXCR4 and CCR7, along with adhesion receptors αvβ3, α6β1, and α4β1, help cells move in the right direction and out of blood vessels. Genomic changes that are typical of aggressive OS, like losing TP53/RB1, amplifying MYC/MDM2, activating KDM6B, and shifting to a glycolytic metabolism, make it easier for the disease to spread. (Right) The lung microenvironment offers a conducive “soil” that is actively influenced by signals from both the host and the tumor. Ezrin–Akt–mTOR signaling, IL11/STAT3 activation, fibroblast-derived IL-11, the LOX family driving ECM remodeling, and platelet-mediated tumor protection are all important immune and stromal mediators. Immunosuppressive elements within the metastatic niche, including M2-like tumor-associated macrophages (TAMs) such as IL-10, TGF-β, and CCL22, myeloid-derived suppressor cells (MDSCs) that inhibit CD8⁺ T-cell function, and the upregulation of PD-L1, promote tumor immune evasion. Increased levels of IL-6 and IL-8 further promote inflammation, angiogenesis, and the spread of cancer. These tumor-intrinsic and microenvironmental mechanisms work together to make OS lung metastasis more effective.
Figure 1. Key molecular drivers and microenvironmental determinants of OS metastasis in the lung. Molecular pathways in OS and lung microenvironmental elements that facilitate metastatic colonization. A combination of tumor-intrinsic signaling pathways, chemokine-integrin axes, and genomic changes drives OS metastasis. These changes make it easier for cancer cells to invade, intravasate, and colonize distant organs, mostly in the lungs. (Left) Signaling molecules that come from tumors, like WNT/β-catenin, MAPK, PI3K/AKT, RAB22A-NEOF fusions, and RhoA, make cells more mobile, help them survive, and make them better at spreading to other parts of the body. Chemokine networks, such as CXCL12/CXCR4 and CCR7, along with adhesion receptors αvβ3, α6β1, and α4β1, help cells move in the right direction and out of blood vessels. Genomic changes that are typical of aggressive OS, like losing TP53/RB1, amplifying MYC/MDM2, activating KDM6B, and shifting to a glycolytic metabolism, make it easier for the disease to spread. (Right) The lung microenvironment offers a conducive “soil” that is actively influenced by signals from both the host and the tumor. Ezrin–Akt–mTOR signaling, IL11/STAT3 activation, fibroblast-derived IL-11, the LOX family driving ECM remodeling, and platelet-mediated tumor protection are all important immune and stromal mediators. Immunosuppressive elements within the metastatic niche, including M2-like tumor-associated macrophages (TAMs) such as IL-10, TGF-β, and CCL22, myeloid-derived suppressor cells (MDSCs) that inhibit CD8⁺ T-cell function, and the upregulation of PD-L1, promote tumor immune evasion. Increased levels of IL-6 and IL-8 further promote inflammation, angiogenesis, and the spread of cancer. These tumor-intrinsic and microenvironmental mechanisms work together to make OS lung metastasis more effective.
Preprints 187387 g001

8.2. Chondrosarcoma (CS)

Chondrosarcoma (CS) is a diverse group of malignant bone tumors characterized by the synthesis of cartilaginous matrix168. It primarily impacts adults and exhibits a preference for the axial skeleton, particularly the pelvis and proximal long bones, including the femur and humerus169. Tumors arise from chondrocytes or their precursors, with most instances classified as conventional central CS170. Less common subtypes, like dedifferentiated and mesenchymal CS, have their own biological traits and are more aggressive in the clinic, which leads to worse outcomes171. The risk of metastasis varies significantly based on histologic subtype and grade, as established by classic studies that highlighted the prognostic significance of morphological characteristics172. In studies involving large populations, traditional CS demonstrates a relatively low metastatic rate, with approximately 6% of patients exhibiting metastases at diagnosis173. On the other hand, dedifferentiated variants and higher-grade lesions are more likely to spread early174175. When metastasis occurs, the lungs are the most common site, followed by the pleura and skeletal sites (ribs, spine). Nodal spread is not very common. The frequent and early dissemination in dedifferentiated and mesenchymal carcinoma signifies the necessity for accurate histological subtyping. Molecular pathogenesis signifies persistent genetic and epigenetic modifications. Mutations in IDH1 and IDH2 happen in about half of CS151 cases. These mutations make the oncometabolite D-2-hydroxyglutarate, which inhibits α-ketoglutarate–dependent dioxygenases and alters DNA and histone methylation176177. Several cohorts have reported that IDH mutation status is associated with enhanced metastasis-free survival, suggesting that alternative molecular pathways facilitate metastasis in IDH-mutant CS178. In addition to IDH, dysregulated Hedgehog signaling and PI3K/AKT activation are present; however, Smoothened inhibition has not produced significant clinical benefit179. Genomic studies have also found changes in COL2A1 (type II collagen) and TP53 in some patients, but we still don’t know exactly what these changes do to help cancer spread180. The CS TME is characterized by a dense cartilaginous extracellular matrix (ECM) abundant in collagen and proteoglycans, which impedes drug delivery and transmits pro-invasive signals181. MMP-1/-2/-13 must change the ECM for invasion to happen, and their levels are linked to how aggressive the cells are182. Simultaneous overexpression of lysyl oxidase (LOX) enhances collagen cross-linking and matrix stiffening, a change that seems contradictory but can actually help migration by increasing integrin-mediated traction183184. CS is generally hypovascular and hypoxic, which stabilizes HIF-1α and starts programs for angiogenesis and migration185. Hypoxia also causes extracellular vesicles and exosomes to be released, which can change the way the immune system works in the area (for example, by changing macrophages to an M2 phenotype) and make an immunosuppressive niche through IL-10 and TGF-β186. Non-malignant stromal components, including mesenchymal stromal cells and fibroblasts, supply growth factors and matrix-modifying enzymes; elevated microvessel density is associated with tumor grade and metastatic potential187. There are many signaling pathways that work together to control growth, survival, invasion, and resistance to treatment. These include PI3K/AKT/mTOR, SRC, and TGF-β188. Dedifferentiated CS frequently accumulates additional genomic abnormalities akin to OS, thereby exacerbating the metastatic phenotype189. Interactions that favor bones, like RANKL signaling, may cause seeding to happen in the skeleton190. Due to the infrequency of the condition, a significant portion of the evidence base is derived from correlative human tissue analyses and in vitro studies. However, the data consistently associate elevated MMP expression, HIF-1α activity, and unique exosomal RNA signatures with metastatic risk186. Proof-of-concept studies indicate that targeting angiogenesis/HIF-1α or macrophage polarization may retard progression191. Anti-angiogenic tyrosine kinase inhibitors (TKIs), such as pazopanib and regorafenib, have demonstrated limited effectiveness in stabilizing disease progression in advanced CS192. Epidemiologically, CS constitutes approximately 20-30% of all malignant bone tumors, ranking as the second most prevalent primary malignant bone neoplasm after OS. CS primarily occurs in adults over 40 years old193, 194, whereas OS is more common in children and adolescents. It is a term that includes groups of things that have different biology, genetics, and epigenetics. The occurrence of CS, especially atypical cartilaginous tumor (ACT), the low-grade variant, has risen, likely due to an aging population and enhanced diagnostic imaging195. Most patients have good outcomes after a wide resection because conventional CS grows slowly and doesn’t spread very often. Nonetheless, advanced, metastatic, or unresectable disease presents a dismal prognosis due to its resistance to chemotherapy and radiotherapy, coupled with a scarcity of effective systemic treatments. Recent studies have underscored a range of genetic and molecular modifications associated with disease advancement and the transition to high-grade or dedifferentiated phenotypes. Some of the most important ones are changes to isocitrate dehydrogenase 1 and 2 (IDH1/2), an increase in EPAS1, which encodes the hypoxia-inducible factor 2-alpha (HIF-2α), and an increase in the SIRT1/HIF-2α signaling axis. These changes make tumors more aggressive, change their metabolism, and help them adapt to low oxygen levels, which leads to malignant transformation and resistance to treatment. It also delineates the progress and constraints of near-patient preclinical models and the potential of novel therapies that target cancer stem cell dependencies or utilize immunological strategies. Integrative profiling has improved risk stratification. A CS multi-omic signature derived from mRNA, microRNA, and DNA methylation identified high-risk patients in one of the largest genetically characterized cohorts to date, highlighting the combined effects of upregulated cell-cycle programs, silencing of the 14q32 imprinted locus (with downregulation of miR-154, miR-382, and miR-384, previously shown to suppress bone sarcoma growth), and genome-wide hypermethylation induced by IDH mutations in driving higher grade and worse prognosis196. This stratification framework delineated three favorable-prognosis subgroups-IDHwt/14q32high, IDHmut/14q32high, and IDHwt/14q32low, alongside two intermediate-risk groups (IDHmut/14q32low and Polifhigh) and a dedifferentiated cohort (14q32low/Prolifhigh) correlated with the most adverse clinical outcomes189. These results highlight the importance of comprehensive molecular profiling beyond IDH mutation status alone and assist in resolving previously noted discrepancies in prognostic classification. Single-cell RNA sequencing identified four signatures based on proliferation, stromal, or leukocyte-related genes. High-grade and dedifferentiated tumors exhibited elevated proliferation indices, with an immunosuppression index distinguishing the dedifferentiated group, while a “active immune response” index characterized low-growing tumors180. The ER-stress regulators DDIT3/CHOP and HSPA5 were recognized as survival indicators in standard central CS, with increased expression linked to adverse outcomes. In CS PDX models, inducing ER stress accelerated growth, while its inhibition hindered progression, establishing ER stress as a therapeutic target. Epigenetic therapies are mechanistically justified due to IDH1/2-induced elevations in DNA and histone methylation; however, IDH-mutated dedifferentiated chronic lymphocytic leukemia (CS) displays reduced hypermethylation and distinct loci in comparison to IDH-mutated conventional CS194. The FDA has approved DNA hypomethylating agents (like 5-aza-2’-deoxycytidine and decitabine) and HDAC inhibitors (like vorinostat, romidepsin, belinostat, and Panobinostat) for use in hematologic malignancies. In preclinical studies, 5-aza plus vorinostat was more effective than either agent alone in vitro and in JJ012 xenografts. It also caused more DNA-damage responses, activated interferon-stimulated genes (including PD-L1), and activated the innate immune system39, 197. A phase II trial of guadecitabine (DNMTi) and belinostat (HDACi) in unresectable or metastatic conventional CS (NCT04340843) failed to achieve the primary overall response rate (ORR) endpoint198. The next steps involve assessing the efficacy of epigenetic therapy in conjunction with immune checkpoint inhibitors and chemotherapy. In general, CS metastasis happens when intrinsic factors (like IDH mutations, COL2A1/TP53 changes, and pathway dysregulation) and extrinsic factors (like ECM remodeling, hypoxia, and immune reprogramming) come together. The fact that IDH-mutant tumors may have better metastasis-free survival highlights molecular heterogeneity and suggests that there are different metastatic circuits in different subgroups180. Current research priorities concentrate on elucidating the metastatic mechanisms associated with IDH–wild-type CS, creating physiologically relevant preclinical models199.

8.3. Liposarcoma (LPS)

Liposarcoma (LPS) is a biologically diverse group of malignant adipocytic tumors that includes many histologic subtypes that differ greatly in their genetic drivers, clinical behavior, and treatment weaknesses200. The primary variants include well-differentiated LPS (WDLS), dedifferentiated LPS (DDLS), myxoid/round-cell LPS (MLPS), and pleomorphic PLPS, each characterized by unique morphology, genetic alterations, and clinical features201. In older adults, WDLS and DDLS are most likely to occur in the deep soft tissues of the extremities or retroperitoneum. These entities illustrate a biological continuum wherein DDLS emerges from pre-existing WDLS via dedifferentiation. MLPS usually shows up in the arms and legs of younger adults and is marked by a lot of myxoid stroma and a branching vascular pattern. Its molecular signature is the FUS-DDIT3 (TLS-CHOP) fusion oncogene produced by the t(12;16)(q13;p11) chromosomal translocation202. PLPS is the rarest but most aggressive subtype, usually found in the limbs, with a high-grade pleomorphic shape and different types of fat cells203. The risk of metastasis varies significantly across subtypes204. WDLS seldom metastasizes, and mortality is generally associated with local recurrence, especially in retroperitoneal tumors where complete resection is difficult. DDLS, on the other hand, is very aggressive and can spread to other parts of the body in up to 30% of cases, usually to the lungs205. MLPS, on the other hand, is more likely to spread outside of the lungs, usually to the bone, spine, and retroperitoneal soft tissues; skeletal metastases happen in about 17% of cases206. PLPS exhibits characteristics akin to undifferentiated pleomorphic sarcoma, including rapid dissemination, pulmonary metastases, and unfavorable survival rates207. Molecular pathogenesis is specific to subtypes. WDLS and DDLS are marked by a steady increase in chromosome 12q13-15, which includes MDM2 (almost always) and CDK4 (often). These two genes work together to cause cancer by turning off p53-mediated checkpoints and speeding up the cell cycle208. The FUS-DDIT3 fusion is what makes MLPS. It stops adipocytic differentiation and causes tumors to grow. Some isoforms are linked to a higher risk of skeletal metastasis209. More mutations, like TP53 and RB1, may build up over time and are linked to resistance to treatment and the ability to spread to other parts of the body210. The TME also changes how metastasis works. DDLS frequently arises within a dense, fibrotic stroma populated by cancer-associated fibroblasts, which remodel the extracellular matrix and facilitate tumor growth211. Exosomes from tumors that carry oncogenic RNAs and microRNAs help form pre-metastatic niches, especially in the lungs212. Immune profiling of DDLS shows that there are inflamed and non-inflamed subtypes. The different types of T-cell infiltration and macrophage polarization affect the prognosis and response to treatment213. The CXCR4-CXCL12 chemokine axis is very important for guiding myxoid LPS cells to secondary organs that are rich in CXCL12. There, ligand–receptor gradients increase the cells’ ability to move, chemotaxis, and survive during dissemination. When tumor-derived angiogenic mediators like VEGF, ANGPT2, and HIF-regulated cytokines are spread around, they cause new blood vessels to form and change the structure of the local stroma to make it easier for metastases to grow214. More generally, receptor tyrosine kinase pathways, such as PDGFR and IGF, along with the activation of PI3K/AKT/mTOR signaling, support proliferation, invasion, and survival across various LPS subtypes215. In clinical practice, these biological differences lead to different ways of keeping an eye on and treating patients. For instance, MLPS patients are now advised to have regular spinal imaging because they are at a high risk of bone metastases216. In DDLS, immune checkpoint blockade has demonstrated restricted effectiveness, highlighting the necessity for predictive biomarkers and combinatorial strategies that focus on TGF-β, angiogenesis, or immune evasion pathways217. The spread of LPS is controlled by a combination of genetic factors that are specific to each subtype, the environment around the cells, and signaling pathways. The different ways that MLPS and DDLS spread show that we need to be very careful when watching and treating them. Future priorities include turning genomic insights into targeted treatments, improving biomarker-driven stratification, and making preclinical models that accurately mimic metastatic niches to help new treatments get developed218. About 1% of all cancerous tumors are sarcomas. LPS, a type of soft tissue sarcoma, is the most common histological type, accounting for 15–20% of cases. LPS arises from adipocytic differentiation and primarily presents in the lower extremities or retroperitoneum219. It is classified according to immunohistochemical profiles, cellular morphology, and related genetic alterations220. Each subtype displays unique biological characteristics, molecular signatures, and pharmacological sensitivities220. Surgery is still the most important part of treating LPS. But the location of the anatomy has a big effect on resectability and prognosis. Retroperitoneal LPS frequently develop asymptomatically within a large cavity, complicating complete resection and leading to a high rate of recurrence. Because having surgery over and over again is hard on the body and mind, how well the first surgery was done is very important for the long-term outcome. Systemic therapy for LPS predominantly utilizes anthracycline-based chemotherapy, sometimes in conjunction with other cytotoxic agents to enhance efficacy, albeit at the cost of heightened toxicity221. Nonetheless, advancements in molecularly targeted and immunotherapeutic strategies are broadening the spectrum of treatment alternatives, most LPS subtypes do not respond well to standard chemotherapies. This review outlines the clinicopathologic characteristics, molecular pathogenesis, and contemporary management strategies of LPS subtypes, with a focus on novel approaches, including targeted therapies and immunotherapies. As our understanding of genetics and molecular biology grows, we can expect that better-designed treatments will lead to better clinical results. MLPS seems to be more sensitive to drugs than other subtypes, which is a good sign for drug development. New drugs, such as anthracycline derivatives, TKIs, marine-derived compounds, and immune modulators, are showing different levels of benefit, which supports the use of combination strategies in the future. Because LPS is rare and can change, it is very important to make an accurate diagnosis, use advanced imaging techniques, and work together with people from different fields. Ultimately, improvements in LPS management will depend on precision medicine techniques and large, multicenter clinical trials to find better treatment options and find a balance between effectiveness and side effects.
Figure 2. Oncogenic drivers and tumor microenvironmental mechanisms underlying malignant melanoma metastasis. Integrated oncogenic programs and tumor microenvironmental cues driving malignant melanoma progression and metastasis. Malignant melanoma progression initiates with deregulation of lineage and EMT-associated transcriptional programs. (Left) Loss of melanocytic differentiation factors such as MITF, along with downregulation of ZEB2 and SNAI2, shifts melanoma cells toward a dedifferentiated, invasive phenotype. Concurrent upregulation of ZEB1, TWIST1, and activation of MAPK, PI3K/AKT, RAS, RAF, and ERK signaling pathways promotes EMT, survival, motility, and therapy resistance. (Center) Key oncogenic drivers include NRAS mutations (Q61R, Q61K, Q61L, Q61H), less common NRAS variants (G12D, G12V, G13R, G13D), BRAFV600E, TERT promoter mutations (C228T/C250T), CDKN2A loss, NF1 loss, and PTEN loss, which collectively amplify proliferation, invasion, metabolic rewiring, and immune evasion. These drivers also facilitate intravasation, survival in circulation, and endothelial transmigration. (Right) The melanoma TME is enriched with CAFs, M2-like macrophages, MDSCs, Tregs, neutrophils, and vascular/lymphatic growth signals (VEGF, TGFβ, IL-10). Immune checkpoint ligands PD-L1, PD-1, TIM-3, LAG-3, and TIGIT induce profound T-cell exhaustion. Proteolytic remodeling via MMP2/MMP9, adhesion changes via N-cadherin, and EMT transcription factors (SNAIL, SLUG, ZEB1) collectively enhance invasion and dissemination. ECM-modifying enzymes including the LOX family, and chemokine axes such as CCL22, CCL17, CXCL12, and IL-6, foster metastatic niche formation. Dysregulated CD8⁺ T cells and chronic inflammation create a permissive environment for metastatic outgrowth in lung, brain, bone, and visceral organs.
Figure 2. Oncogenic drivers and tumor microenvironmental mechanisms underlying malignant melanoma metastasis. Integrated oncogenic programs and tumor microenvironmental cues driving malignant melanoma progression and metastasis. Malignant melanoma progression initiates with deregulation of lineage and EMT-associated transcriptional programs. (Left) Loss of melanocytic differentiation factors such as MITF, along with downregulation of ZEB2 and SNAI2, shifts melanoma cells toward a dedifferentiated, invasive phenotype. Concurrent upregulation of ZEB1, TWIST1, and activation of MAPK, PI3K/AKT, RAS, RAF, and ERK signaling pathways promotes EMT, survival, motility, and therapy resistance. (Center) Key oncogenic drivers include NRAS mutations (Q61R, Q61K, Q61L, Q61H), less common NRAS variants (G12D, G12V, G13R, G13D), BRAFV600E, TERT promoter mutations (C228T/C250T), CDKN2A loss, NF1 loss, and PTEN loss, which collectively amplify proliferation, invasion, metabolic rewiring, and immune evasion. These drivers also facilitate intravasation, survival in circulation, and endothelial transmigration. (Right) The melanoma TME is enriched with CAFs, M2-like macrophages, MDSCs, Tregs, neutrophils, and vascular/lymphatic growth signals (VEGF, TGFβ, IL-10). Immune checkpoint ligands PD-L1, PD-1, TIM-3, LAG-3, and TIGIT induce profound T-cell exhaustion. Proteolytic remodeling via MMP2/MMP9, adhesion changes via N-cadherin, and EMT transcription factors (SNAIL, SLUG, ZEB1) collectively enhance invasion and dissemination. ECM-modifying enzymes including the LOX family, and chemokine axes such as CCL22, CCL17, CXCL12, and IL-6, foster metastatic niche formation. Dysregulated CD8⁺ T cells and chronic inflammation create a permissive environment for metastatic outgrowth in lung, brain, bone, and visceral organs.
Preprints 187387 g002

8.4. Melanoma

Melanoma is a highly aggressive malignancy originating from neural crest-derived melanocytes found in cutaneous, mucosal, and ocular regions; cutaneous melanoma is the predominant subtype222. Histologically, lesions range from radial growth-phase tumors limited to the epidermis to vertically invasive tumors that penetrate the basement membrane; cells may be pigmented or amelanotic, but the hallmark is invasive growth beyond the site of origin223. Melanoma is clinically significant because it spreads to other parts of the body early and often through both lymphatic and hematogenous pathways, which explains why it has such a big effect on skin cancer deaths224. The initial phase of dissemination typically entails migration via dermal lymphatics to regional lymph nodes; consequently, nodal status serves as a crucial prognostic indicator, informing staging and management225. After that, the disease spreads through the blood to the lungs, liver, brain, bones, and skin/subcutaneous tissue. Brain metastases happen in a lot of advanced cases and have a big effect on survival and quality of life226. In-transit metastasis, in which tumor deposits form between the primary site and the regional basin, is a unique sign of lymphatic dissemination. This is because melanoma has lymphotropism and is hard to treat because it spreads to multiple places227. At the molecular level, metastatic competence is based on well-defined oncogenic changes that occur repeatedly228. BRAF V600 mutations (most often V600E) turn on MAPK/ERK signaling all the time and are found in about half of cutaneous melanomas. NRAS mutations (about 15–20%) activate both the MAPK and PI3K/AKT pathways229. Events that cause tumor suppressors such as NF1, PTEN, and CDKN2A to stop functioning make growth and survival signaling even less stable230. Highly prevalent TERT promoter mutations that increase telomerase activity and cellular immortality are other factors that accelerate progression231. Melanoma cells use the MAPK, PI3K/AKT, WNT/β-catenin, and NF-κB signaling pathways to control growth, survival, immune evasion, and metastasis232. Invasion and dissemination rely on dynamic changes in adhesion and motility. Melanoma cells frequently downregulate E-cadherin and upregulate N-cadherin, facilitating detachment from keratinocyte constraints and enhancing interactions with stromal and endothelial components233. MMP-2 and MMP-9 promote the degradation of the basement membrane and extracellular matrix (ECM), whereas integrins, especially αvβ3 and α4β1, facilitate migration through various tissue matrices234. Cells can change their shape from mesenchymal to amoeboid in response to changes in their microenvironment235. Chemokine axes, like CCR4/CCR10 for skin homing and CCR7 for nodal migration, help direct organotropism. This is similar to how ligands are expressed in target tissues236. The TME determines how well a cancer spreads and how well it responds to treatment. Fibroblasts associated with cancer alter the ECM (fibronectin, collagens) and give cells signals to stay alive and move237. Tumor-associated macrophages often exhibit an M2-like phenotype, secreting VEGF, TGF-β, and IL-10 that facilitate angiogenesis, immune suppression, and invasion238. The immune landscape is critical for prognosis: a high number of CD8⁺ T cells in the right places is a good sign, while a high number of regulatory T cells and M2 macrophages is a bad sign239. Melanoma promotes lymphangiogenesis in lymph nodes, enlarging lymphatic pathways for dissemination240. Immune evasion is crucial for successful metastasis. Upregulation of PD-L1 on melanoma cells activates PD-1 on T cells, impairing effector function and facilitating tumor progression, a pathway now effectively targeted in clinical settings241. The “immunotherapy revolution” underscores the TME’s importance: checkpoint inhibitors have changed the lives of many patients, but primary and acquired resistance remain common and require biomarker-guided combinations242. Preclinical models closely resemble human disease and have elucidated the biology of metastasis and therapeutic resistance. Genetically engineered mouse models, like BRAFV600E with PTEM-/-, mimic spontaneous metastasis. B16 syngeneic and human xenograft models build on these results, especially for lung/brain spread and treatment evaluation243. Immune infiltration patterns and driver genotypes correlate with prognosis and therapeutic response, elucidating prevalent resistance mechanisms via secondary mutations or pathway reconfiguration244.Overall, melanoma metastasis results from the interaction of well-known oncogenic drivers (such as mutations in BRAF, NRAS, and the TERT promoter), changes in adhesion and motility that help the cancer spread, chemokine-directed organotropism, and an immune-modulatory TME245. More investigation is needed to verify the factors influencing immune-infiltration heterogeneity, developing predictive biomarkers for targeted and immunotherapies, and devising logical combinations that concurrently tackle tumor-intrinsic pathways and TME-mediated resistance246. Single-cell/spatial multi-omics, liquid biopsy monitoring, and next-generation, immune-competent preclinical models that better mimic human metastatic disease247 will speed up progress.

8.5. Hepatocellular Carcinoma (HCC)

Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer, originates from hepatocytes, usually in association with chronic liver disease and cirrhosis248. Most cases arise from chronic hepatic inflammation caused by hepatitis B or C infection, alcohol-related liver disease, or non-alcoholic steatohepatitis249. Histologically, HCC exhibits trabecular and pseudoglandular proliferation of atypical hepatocytes accompanied by architectural distortion. Fibrolamellar HCC is a unique type that happens in younger patients who don’t have cirrhosis. It has its own molecular characteristics and clinical behavior250. Clinically, HCC is widespread worldwide and is marked by aggressive biology and early metastatic potential, leading to unfavorable survival outcomes251. HCC metastasis indicates its intrahepatic origin and significant vascular invasiveness into the portal and hepatic veins, which promote hematogenous dissemination and are associated with a poor prognosis252. The lung is the most common place outside the liver, followed by the lymph nodes in the abdomen, and the spread of cancer through the peritoneum. Bone metastases, which usually involve the axial skeleton, often cause severe pain and fractures253. Other targets include the diaphragm, brain, adrenal glands, and pleural surfaces253. Extensive clinical studies consistently indicate a decreasing prevalence of involvement in the lung, peritoneum, bone, spleen, adrenal glands, brain, pleura, and kidneys254. Brain metastases are infrequent but generally occur late and indicate a poor prognosis for survival255. At the molecular level, chronic inflammatory injury creates a mutagenic environment that promotes cancer growth and the spread of cancer cells to other parts of the body256. Recurrent alterations include TERT promoter mutations that reactivate telomerase257, TP53 mutations that hinder genomic surveillance, and CTNNB1 (β-catenin) mutations that promote Wnt pathway activation 258259. Other factors that sustain angiogenesis and promote metastatic colonization260 include MET activation and widespread upregulation of the VEGF pathway. Metastatic dissemination entails epithelial–mesenchymal transition, facilitated by Snail/Zeb factors and often triggered by TGF-β-rich inflammation, while matrix metalloproteinases, including MMP-9, degrade the extracellular matrix to permit invasion261. HCC develops in a microenvironment characterized by cirrhosis, fibrosis, and inflammation, which is intrinsically immunosuppressive262. Activated hepatic stellate cells and portal fibroblasts generate a collagen-rich matrix and induce tissue stiffening, which influences tumor behavior and hinders drug penetration263. CAFs alter ECM, release VEGF, PDGF, and chemokines, and keep the immune system out by signaling through IL-6 and TGF-β and building physical barriers264. Macrophages, particularly M2-like subsets, play roles in immune suppression, angiogenesis, and matrix remodeling265. VEGF-induced endothelial hyperproliferation results in disordered vasculature that promotes intravasation and is associated with vascular invasion266. These interactions between stromal and immune cells cooperate with oncogenic pathways such as PI3K/AKT activation, PTEN loss, FGF/IGF signaling, and PD-L1 upregulation to sustain tumor growth, invasion, and evasion of the immune system260267268. Molecular profiling has elucidated metastasis-associated signatures, including exosome-derived miRNAs that influence distant microenvironments and promote the formation of metastastatic niches269. In clinical practice, multiple-organ metastases frequently occur after surgical resection, with the lung, bone, and peritoneum being the most commonly affected sites270. In general, HCC metastasis is driven by chronic inflammation-induced mutagenesis, including mutations in TERT, TP53, and CTNNB1.

8.6. Glioblastoma (GBM)

GBM is the most aggressive primary cancer of the central nervous system. The 2021 update of the World Health Organization classification274 states that it is a WHO grade IV astrocytic tumor. GBM, which mainly arises from glial cells, primarily astrocytes, is characterized by pseudopalisading necrosis and microvascular proliferation under a microscope. These traits indicate a malignant biology and set it apart from lower-grade gliomas275. In clinical settings, GBM exhibits diffuse infiltration, rapid recurrence, and unfavorable outcomes despite aggressive multimodal treatment, including maximal resection, radiotherapy, and temozolomide chemotherapy, with median survival rarely exceeding 15 months276. Unlike most systemic malignancies, GBM infrequently metastasizes beyond the central nervous system. This unique confinement is due to the protective blood-brain barrier, the absence of a traditional lymphatic system, the specialized neural microenvironment, and the short clinical course that limits the time for systemic dissemination277. As a result, GBM “metastasis” is characterized not by distant organ colonization but by significant local invasion within the brain parenchyma. Tumor cells invade through white matter tracts, perivascular spaces, and cerebrospinal fluid pathways, sometimes resulting in leptomeningeal or spinal “drop” metastases278. Rare instances of extracranial dissemination are generally associated with surgical procedures, such as craniotomy or ventricular shunting, which may facilitate cellular egress277. This pattern of diffuse invasion within the brain, rather than hematogenous dissemination, characterizes the lethal persistence of GBM. The molecular composition of GBM is highly heterogeneous. Verhaak et al. (2010)279 showed that transcriptomic profiling has identified three main molecular subtypes. Each of these subtypes is associated with specific driver changes. The classical subtype is distinguished by the amplification and mutation of the epidermal growth factor receptor (EGFR), often involving the constitutively active EGFRvIII variant. This change keeps the Ras/MAPK and PI3K/AKT pathways active all the time, which makes cells grow and spread more easily280. The mesenchymal subtype is often characterized by NF1 loss and activation of TGF-β signaling, leading to an EMT-like, highly invasive phenotype rich in inflammatory and angiogenic programs281. The proneural subtype is usually characterized by either PDGFRA amplification or IDH1 mutations. The latter is a sign of secondary GBMs that develop from lower-grade gliomas and have a CpG island methylator phenotype (G-CIMP) with a relatively good prognosis276. This classification highlights that GBM is not a singular disease but rather a spectrum of molecularly distinct entities characterized by convergent features of unregulated proliferation, invasion, and therapeutic resistance. Integrin-mediated adhesion and remodeling of the extracellular matrix control invasion. For instance, α6β1-laminin interactions help cells move around blood vessels, and matrix metalloproteinases like MMP-2 and MMP-9 break down type IV collagen and change the brain’s extracellular matrix282. Glioma cells demonstrate motility plasticity, transitioning between elongated, protease-dependent mesenchymal migration and rounded, deformable amoeboid movement283. This flexibility allows it to spread through both dense and flexible parts of the brain. Hypoxia further increases invasiveness by stabilizing HIF-1α, which in turn increases VEGF and EMT transcription factors like ZEB1. This leads to angiogenesis and mesenchymal transition284. Tumor-associated macrophages and microglia reportedly comprise up to fifty percent of the tumor mass and frequently exhibit an M2-like phenotype, secreting IL-10 and TGF-β to inhibit T-cell activity238. Single-cell analyses have elucidated a range of tumor-associated macrophage (TAM) states and their reciprocal interactions with glioma cells that facilitate invasion and angiogenesis285. On the other hand, tumor-infiltrating lymphocytes are few and mostly worn out, which makes immune checkpoint blockade less effective286. Reactive astrocytes and endothelial cells also help by releasing growth factors and cytokines that make a tumor-supportive niche287. The outcome is a microenvironment that protects the tumor from immune destruction and promotes ongoing growth. Genetically, GBM is caused by the wrong activation of the receptor tyrosine kinase and the signaling pathways that follow it. EGFR amplification, PTEN loss, and TP53 mutation are among the most common changes288. EGFR activation continuously activates the MAPK and PI3K/AKT signaling pathways, which help cells grow, prevent cell death, and adapt to changes in metabolism289. The PI3K/AKT/mTOR pathway is modified in up to 86% of GBMs and serves as a crucial nexus for survival and metabolic control. Simultaneously, mTORC2-mediated AKT phosphorylation enhances invasive and therapy-resistant phenotypes290. The interactions among the MAPK, PI3K/mTOR, and PKC pathways form a robust signaling network that sustains tumor growth. The MNK-dependent phosphorylation of eIF4E for translational control is another place where things come together. Tomivosertib, an MNK inhibitor, prevents eIF4E activation, slowing angiogenesis and making tumors more sensitive to temozolomide. This shows that translational regulation is a therapeutic weakness that can be exploited291. Epigenetic dysregulation plays a major role in the diversity and adaptability of GBM. Phenotypic plasticity, recognized as a nascent characteristic of cancer in GBM, is profoundly associated with epigenetic reprogramming292. Patterns of DNA methylation affect both the biology of tumors and how well treatments work. For example, methylation of the MGMT promoter predicts a good response to temozolomide, and the G-CIMP phenotype linked to IDH1 mutations defines a unique epigenetic and prognostic subgroup293. At the chromatin level, EZH2-mediated H3K27 methylation inhibits tumor suppressors like PTEN and collaborates with PI3K signaling to promote oncogenic transcription294. By deacetylating α-tubulin and stabilizing EGFR295, HDAC6 helps glioma cells grow and move. Pharmacological inhibition of HDAC6 or EZH2 diminishes invasiveness in preclinical models. Nonetheless, clinical trials involving broad HDAC inhibitors or DNA demethylating agents have thus far demonstrated limited efficacy, highlighting the necessity for more selective, brain-penetrant epigenetic modulators. Resistance to therapy in GBM is due to a combination of genetic variation, stem-like plasticity, and protection from the microenvironment296. Recurrence usually happens because of resistant subclones that were already there and survived chemoradiation. Recurrent tumors often show mesenchymal enrichment and changes induced by therapy297. Glioma stem-like cells remain dormant, facilitate DNA repair by activating ATM/ATR, and replenish tumors post-therapy298. These cells also increase the activity of efflux transporters and anti-apoptotic genes, which makes multidrug resistance even stronger. These mechanisms elucidate the reasons for the recurrent failures of targeted therapies targeting single pathways. EGFR inhibitors and anti-angiogenic agents like bevacizumab produce temporary responses but lack a sustained survival advantage299. Recent progress is changing the way we treat diseases. Researchers are looking into new ways to get drugs into the body, such as convection-enhanced drug infusion, nanoparticles, and peptide-mediated blood–brain barrier penetration300. Non-pharmacologic modalities, especially tumor-treating fields, have improved median survival when combined with temozolomide, underscoring the importance of integrating novel technologies301. PD-1 blockade, multi-antigen vaccines, and regionally administered CAR T cells that target IL13Rα2 or HER2, can trigger an anti-tumor immune response even in the immunosuppressive environment of GBM302. Oncolytic virotherapy has demonstrated potential; the HSV-1-derived Delytact (G47Δ) attained sustained disease control in a cohort of patients with recurrent GBM303, representing the inaugural approved virotherapy for malignant glioma. Even with these small improvements, GBM is still a model of an adaptive cancer. Its confinement in the brain conceals a remarkable capacity to exploit the microenvironment, evade treatment, and recur. For future success, we will need to use combinations of treatments that target oncogenic signaling, epigenetic plasticity, and immune suppression simultaneously, while also making it easier for drugs to cross the blood-brain barrier. Combining genomic, epigenomic, and spatial single-cell analyses is helping us learn more about how different types of cells in a tumor work together and how to develop combination therapies304. With ongoing progress in multi-omic profiling, immunoengineering, and precision drug delivery, the enduring therapeutic stalemate of GBM may finally commence to dissolve.
Figure 3. Integrated oncogenic signaling, microenvironmental factors, and resistance mechanisms in GBM. Characteristics of GBM biology encompass its restricted systemic metastasis, oncogenic drivers, microenvironmental composition, and the mechanisms that contribute to therapeutic resistance. (Top left) GBM rarely metastasizes beyond the central nervous system owing to the prohibitive blood-brain barrier, the absence of standard lymphatic drainage, and a neural microenvironment that constrains tumor cell spread. The brief clinical progression of GBM and physiological limitations that hinder systemic spread are additional factors. (Middle left—GBM resistance mechanisms) Therapeutic resistance is caused by a lot of different types of cells in the tumor, the presence of therapy-resistant glioma stem-like cells (GSCs), and an extracellular matrix that is rich in hyaluronic acid and poor in collagen, which protects tumor cells from drugs. Resistance is bolstered by the methylation status of the MGMT promoter, transitions in the mesenchymal state (MES), metabolic reprogramming, and adaptive pathways driven by hypoxia. (Bottom left: GBM microenvironment) The GBM TME is made up of many different types of cells that help tumors grow and suppress the immune system. These include GSCs, tumor-associated macrophages (TAMs), tumor-associated microglia, reactive astrocytes, exhausted T cells, myeloid-derived suppressor cells (MDSCs), and endothelial cells/pericytes. These parts work together to cause invasion, immune evasion, and local recurrence. (Right—Oncogenic signaling) PTEN loss, NF1 loss, EGFR/EGFRvIII activation, PDGFRα amplification, CDKN2A deletion, TERT promoter mutations, EZH2 upregulation, and PDGFRA/IDH1-mutant pathways are the main causes of GBM. These molecular events trigger standard receptor tyrosine kinase (RTK) signaling pathways, such as the PI3K–AKT–mTORC1/2 axis and the EGFR–RAS–RAF–MEK–ERK cascade. This signaling pathway increases the levels of HIF-1α, which in turn increases VEGF-mediated angiogenesis, leading to faster tumor growth and resistance to treatment.
Figure 3. Integrated oncogenic signaling, microenvironmental factors, and resistance mechanisms in GBM. Characteristics of GBM biology encompass its restricted systemic metastasis, oncogenic drivers, microenvironmental composition, and the mechanisms that contribute to therapeutic resistance. (Top left) GBM rarely metastasizes beyond the central nervous system owing to the prohibitive blood-brain barrier, the absence of standard lymphatic drainage, and a neural microenvironment that constrains tumor cell spread. The brief clinical progression of GBM and physiological limitations that hinder systemic spread are additional factors. (Middle left—GBM resistance mechanisms) Therapeutic resistance is caused by a lot of different types of cells in the tumor, the presence of therapy-resistant glioma stem-like cells (GSCs), and an extracellular matrix that is rich in hyaluronic acid and poor in collagen, which protects tumor cells from drugs. Resistance is bolstered by the methylation status of the MGMT promoter, transitions in the mesenchymal state (MES), metabolic reprogramming, and adaptive pathways driven by hypoxia. (Bottom left: GBM microenvironment) The GBM TME is made up of many different types of cells that help tumors grow and suppress the immune system. These include GSCs, tumor-associated macrophages (TAMs), tumor-associated microglia, reactive astrocytes, exhausted T cells, myeloid-derived suppressor cells (MDSCs), and endothelial cells/pericytes. These parts work together to cause invasion, immune evasion, and local recurrence. (Right—Oncogenic signaling) PTEN loss, NF1 loss, EGFR/EGFRvIII activation, PDGFRα amplification, CDKN2A deletion, TERT promoter mutations, EZH2 upregulation, and PDGFRA/IDH1-mutant pathways are the main causes of GBM. These molecular events trigger standard receptor tyrosine kinase (RTK) signaling pathways, such as the PI3K–AKT–mTORC1/2 axis and the EGFR–RAS–RAF–MEK–ERK cascade. This signaling pathway increases the levels of HIF-1α, which in turn increases VEGF-mediated angiogenesis, leading to faster tumor growth and resistance to treatment.
Preprints 187387 g003

8.7. Breast Tumor

Breast cancer metastasis is a very slow and complicated process that only a small number of tumor cells can complete305. These metastasis-initiating cells (MICs) display stem-like characteristics and exceptional phenotypic plasticity, allowing them to endure systemic stresses, circumvent immune responses, and acclimatize to foreign tissue environments306. The induction of epithelial-mesenchymal transition (EMT) is a key factor in MIC plasticity. In breast tumors, transcription factors such as SNAIL, ZEB1, and PRRX1 control this process. These EMT-TFs inhibit epithelial markers (e.g., E-cadherin) and stimulate mesenchymal genes, imparting motility and stemness to cells306. The absence of the EMT inducer Prrx1 has been shown to facilitate MET and metastatic colonization in breast models, underscoring the dynamic and reversible nature of the EMT program307. The tumor microenvironment (TME) has a big effect on EMT activation in breast cancer. For example, cancer-associated fibroblasts (CAFs) secrete TGF-β1 and other substances that induce EMT308. On the other hand, TAMs release cytokines like CCL18 and IL-6 that help cells invade and spread309. For example, CCL18 from TAM starts a signaling cascade through PITPNM3/ANXA2 that turns on PI3K/Akt/GSK3β signaling and raises the level of Snail310. In the same way, IL-6 secreted by TAMs activates the JAK2/STAT3 pathway in breast cancer cells, increasing EMT-TF expression and conferring stem-like, therapy-resistant characteristics55. These microenvironmental signals work with cancer-intrinsic EMT programs to produce cells that are highly mobile and resistant to treatment. To leave the primary site, breast cancer cells need to break through the surrounding stroma and basement membranes. They do this mostly by increasing the levels of matrix metalloproteinases (MMPs), especially MMP-2, MMP-9, and the membrane-anchored MT1-MMP (MMP-14), which break down type IV collagen and other ECM components311. High levels of MMP-2/9 activity are strongly linked to invasive behavior and the spread of cancer to other parts of the body. MT1-MMP, on the other hand, is a key effector that activates pro-MMP-2 to boost proteolytic cascades at the invasive front312. Once in circulation, breast cancer cells endure shear stress and immune surveillance by moving as circulating tumor cell (CTC) clusters. These clusters are about 23-50 times more likely to spread than single CTCs313. Platelet “cloaking” of circulating tumor cells (CTCs) increases the ability of the immune system to avoid detection while also promoting microvascular arrest and vascular loading314. Hypoxic conditions enhance this process by increasing CTC-platelet aggregation, thereby creating a temporary protective niche during hematologenous transit315. After extravasation, disseminated tumor cells (DTCs) that spread to distant organs, such as the lungs and additional bone marrow, can enter a dormancy bottleneck, where they can remain dormant for years. Microenvironmental remodeling often triggers reactivation from dormancy. For example, the neutrophil extracellular trap (NET)-mediated cleavage of laminin produces bioactive fragments that activate b1-integrin-YAP signaling, ultimately promoting metastatic outgrowth316. Organ-specific “seed and soil” interactions control metastatic growth. Breast cancer exhibits specific levels of COX2 (PTGS2) and MMP1, which facilitate vascular remodeling and extravasation317. In brain metastasis, breast cancer cells aberrantly express ST6GALNAC5, facilitating their penetration of the blood-brain barrier318. Blocking COX2 or MMP1 stops lung metastasis, and knocking down ST6GALNAC5 stops brain colonization. Invasive lobular carcinoma (ILC), characterized by the loss of E-cadherin, demonstrates distinct metastatic pathways and increased susceptibility to IGF1R pathway inhibition319. Metastatic colonies change how they use energy in different organs. Breast cancer cells that prefer bone tissue depend on glycolysis and lactate-driven osteolytic remodeling320. Brain metastases adapt to a glutamine-rich environment by enhancing glutamine anaplerosis and metabolic plasticity321. EMT-high cells exhibit a distinct susceptibility: ZEB1-mediated ferroptosis sensitivity via the repression of SCD1 and the accumulation of poly- unsaturated lipids322. New ways to treat people use these ideas. Blocking both TGF-β and CD73 reverses EMT and improves the efficacy of immunotherapy323. Inducing ferroptosis is another way to target cells that initiate metastasis by driving epithelial-mesenchymal transition (EMT)324. ENPP1, an enzyme that breaks down cGAMP, promotes metastasis to the brain and bones and stops STING signaling. It is a promising target for treatment325. Hedgehog-GLI signaling facilitates bone colonization and resistance to endocrine therapy, while SMO/GLI inhibitors have demonstrated preclinical efficacy326. A more in-depth understanding of how these processes work is now being combined with new treatments targeting EMT circuits, metabolic weaknesses such as ferroptosis, pro-metastatic enzymes such as ENPP1, and developmental pathways such as Hedgehog327. This gives hope for breast cancer-specific treatments that can get rid of dormant seeds and stop the deadly spread of metastases327. The ongoing challenge is to turn these discoveries into safe and effective treatments that stop the spread of cancer and improve survival in advanced breast cancer.
Figure 4. Oncogenic drivers, receptor profiles, and organ-specific metastatic pathways in various breast cancer subtypes. Combined oncogenic mechanisms and organotropism for metastasis in major subtypes of breast cancer. Breast tumors are characterized by their receptor status, specifically the expression patterns of ER, PR, HER2, and HR, which influence lineage identity, oncogenic dependencies, and metastatic pathways. (Left) Key oncogenic drivers consist of the amplification or activation of HER2/ERBB2, EGFR, FGFR1/2, and MET; mutations in BRCA1/2, PALB2, and ESR1; activating alterations in KRAS, NRAS, BRAF, PIK3CA, and AKT1; and the loss of PTEN. Other factors that play a role are cell-cycle deregulation (CCND1 amplification, CDK4/6 activation, RB1 loss), EMT regulators (SNAIL, ZEB1, TWIST, PRRX1), and epigenetic modifiers (EZH2, DNMT1, KDM6A/B, HDACs). These changes work together to help tumors survive, change shape, invade, and spread to other parts of the body. (Right) Different types of breast cancer have different ligand, cytokine, and adhesion profiles that control organotropic metastasis: The luminal HER2 subtype has more IGF1, PGE2, TGFβ, PDGF, FGF2, IL-11, IL-1, IL-6, PTHrP, OPN, and RANKL-RANK, which helps bone-trophic signaling. HER2-enriched ER⁺ / Luminal B/ Luminal-HER2 tumors express CXCR4/CXCL12, IL-6, α2β1, α5β1, N-cadherin, LOX, OPN, VEGF, TWIST, and WNT, which makes it easier for them to spread to the liver and bones. L1CAM, COX2, Ang2, IL-8, MMP-1/2/8/9, ST6GALNAC5, VEGF, HBEGF, CXCR4, and CK5 are all present in HER2-enriched Luminal-HER2 and TN-nonbasal basal-like tumors. These markers help the tumors spread to the lungs and brain quickly. TN-nonbasal basal-like Luminal B and HER2⁺/HR⁻/p53-mutant tumors exhibit pronounced inflammatory and proteolytic profiles (TGFβ, EREG, VEGF, MMP-1/2, COX2, LOX), facilitating visceral and pulmonary metastasis. Luminal-type HER2-enriched tumors increase the levels of VEGF-C/VEGF-D, PROX1, LYVE1, and the kallikrein family (KLK10, KLK11, KLK12, KLK13). This shows that they have lymphangiogenic and HER2-driven metastatic traits.
Figure 4. Oncogenic drivers, receptor profiles, and organ-specific metastatic pathways in various breast cancer subtypes. Combined oncogenic mechanisms and organotropism for metastasis in major subtypes of breast cancer. Breast tumors are characterized by their receptor status, specifically the expression patterns of ER, PR, HER2, and HR, which influence lineage identity, oncogenic dependencies, and metastatic pathways. (Left) Key oncogenic drivers consist of the amplification or activation of HER2/ERBB2, EGFR, FGFR1/2, and MET; mutations in BRCA1/2, PALB2, and ESR1; activating alterations in KRAS, NRAS, BRAF, PIK3CA, and AKT1; and the loss of PTEN. Other factors that play a role are cell-cycle deregulation (CCND1 amplification, CDK4/6 activation, RB1 loss), EMT regulators (SNAIL, ZEB1, TWIST, PRRX1), and epigenetic modifiers (EZH2, DNMT1, KDM6A/B, HDACs). These changes work together to help tumors survive, change shape, invade, and spread to other parts of the body. (Right) Different types of breast cancer have different ligand, cytokine, and adhesion profiles that control organotropic metastasis: The luminal HER2 subtype has more IGF1, PGE2, TGFβ, PDGF, FGF2, IL-11, IL-1, IL-6, PTHrP, OPN, and RANKL-RANK, which helps bone-trophic signaling. HER2-enriched ER⁺ / Luminal B/ Luminal-HER2 tumors express CXCR4/CXCL12, IL-6, α2β1, α5β1, N-cadherin, LOX, OPN, VEGF, TWIST, and WNT, which makes it easier for them to spread to the liver and bones. L1CAM, COX2, Ang2, IL-8, MMP-1/2/8/9, ST6GALNAC5, VEGF, HBEGF, CXCR4, and CK5 are all present in HER2-enriched Luminal-HER2 and TN-nonbasal basal-like tumors. These markers help the tumors spread to the lungs and brain quickly. TN-nonbasal basal-like Luminal B and HER2⁺/HR⁻/p53-mutant tumors exhibit pronounced inflammatory and proteolytic profiles (TGFβ, EREG, VEGF, MMP-1/2, COX2, LOX), facilitating visceral and pulmonary metastasis. Luminal-type HER2-enriched tumors increase the levels of VEGF-C/VEGF-D, PROX1, LYVE1, and the kallikrein family (KLK10, KLK11, KLK12, KLK13). This shows that they have lymphangiogenic and HER2-driven metastatic traits.
Preprints 187387 g004

9. Emerging Therapeutic Strategies for Metastatic Disease

Therapeutic advancements in metastatic cancer increasingly embody a mechanistic understanding of tumor evolution, immune evasion, and microenvironmental reconfiguration. Targeted inhibitors that block oncogenic drivers like EGFR and ALK have changed how metastatic lung cancer is treated by selectively breaking up signaling networks that keep cells growing328. This precision paradigm acknowledges that metastasis is regulated by distinct biological programs, such as epithelial-mesenchymal transition (EMT), niche formation, and immune suppression, which can be targeted with specific interventions329. Immune checkpoint inhibitors (ICIs) are still very important for melanoma and lung cancer because they help T-cells work again330. However, many metastatic lesions remain “cold,” meaning they lack dendritic-cell priming and T-cell infiltration. Combination immunotherapies aim to overcome resistance: therapeutic cancer vaccines enhance neoantigen recognition and work in combination with PD-1 blockade331, while CD40 and OX40 agonists boost dendritic-cell activation and T-cell costimulation332. Adoptive cell therapies improve immune system benefits. TIL therapy causes long-lasting responses in melanoma that doesn’t respond to other treatments, and “armored” CAR-T cells that secrete IL-12 or IL-15 fight against solid tumor microenvironments that suppress the immune system333. Furthermore, CAR-macrophages represent an innovative frontier, reprogramming M2 macrophages into inflammatory M1 states and diminishing metastatic lesions in preclinical models334. It is just as important to target the “soil” where the cancer spreads. Stromal-directed agents like PEGPH20 break down hyaluronan, making tumors less dense and allowing drugs to reach them better336. FAP-targeted therapies, on the other hand, break up fibroblast-rich metastatic niches337. In bone metastasis, the inhibition of RANKL by denosumab diminishes skeletal events and decelerates progression338. Inhibition of CXCR4339 or blockade of LOX/LOXL2 to stop ECM stiffening340 are both promising ways to stop pre-metastatic niche formation. Simultaneous advancements in monitoring and delivery are transforming clinical management. Liquid biopsies allow for real-time tracking of how metastatic cancer is changing. The dynamics of ctDNA can predict when the cancer will come back sooner than imaging341, and help doctors choose adaptive therapy methods that change treatment based on how the tumor is changing. Nanomedicine improves the delivery of drugs to metastatic lesions342, and biomaterial-based depot systems like injectable immunostimulatory hydrogels provide long-lasting immune activation in an area to stop the disease from coming back343. New cellular technologies, such as CRISPR-edited T cells344, tumor-homing MSC-based therapies345, and universal iPSC-derived NK/T cells346, hold the potential to provide new modular, off-the-shelf treatments for metastatic disease. These changes show a shift from broad cytotoxic strategies to more precise ones that combine targeted oncogenic blockade. This changing framework sees metastatic cancer as a disease that can be managed over the long term through coordinated therapeutic pressure, rather than as a disease that always leads to death.

Funding

This research received no external funding.

Acknowledgments

Figures in this manuscript were created with BioRender (BioRender.com).

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Fidler IJ. The pathogenesis of cancer metastasis: the ‘seed and soil’ hypothesis revisited. Nat Rev Cancer. 2003;3(6):453-8. PubMed PMID: 12778135. [CrossRef]
  2. Quail DF, Joyce JA. Microenvironmental regulation of tumor progression and metastasis. Nat Med. 2013;19(11):1423-37. PubMed PMID: 24202395; PMCID: PMC3954707. [CrossRef]
  3. Yates LR, Gerstung M, Knappskog S, Desmedt C, Gundem G, Van Loo P, Aas T, Alexandrov LB, Larsimont D, Davies H, Li Y, Ju YS, Ramakrishna M, Haugland HK, Lilleng PK, Nik-Zainal S, McLaren S, Butler A, Martin S, Glodzik D, Menzies A, Raine K, Hinton J, Jones D, Mudie LJ, Jiang B, Vincent D, Greene-Colozzi A, Adnet PY, Fatima A, Maetens M, Ignatiadis M, Stratton MR, Sotiriou C, Richardson AL, Lonning PE, Wedge DC, Campbell PJ. Subclonal diversification of primary breast cancer revealed by multiregion sequencing. Nat Med. 2015;21(7):751-9. Epub 20150622. PubMed PMID: 26099045; PMCID: PMC4500826. [CrossRef]
  4. Turajlic S, Swanton C. Metastasis as an evolutionary process. Science. 2016;352(6282):169-75. PubMed PMID: 27124450. [CrossRef]
  5. Lawson DA, Bhakta NR, Kessenbrock K, Prummel KD, Yu Y, Takai K, Zhou A, Eyob H, Balakrishnan S, Wang CY, Yaswen P, Goga A, Werb Z. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature. 2015;526(7571):131-5. Epub 20150923. PubMed PMID: 26416748; PMCID: PMC4648562. [CrossRef]
  6. Aguirre-Ghiso JA. Models, mechanisms and clinical evidence for cancer dormancy. Nat Rev Cancer. 2007;7(11):834-46. PubMed PMID: 17957189; PMCID: PMC2519109. [CrossRef]
  7. Manjili MH. Tumor Dormancy and Relapse: From a Natural Byproduct of Evolution to a Disease State. Cancer Res. 2017;77(10):2564-9. PubMed PMID: 28507050; PMCID: PMC5459601. [CrossRef]
  8. Sosa MS, Bragado P, Aguirre-Ghiso JA. Mechanisms of disseminated cancer cell dormancy: an awakening field. Nat Rev Cancer. 2014;14(9):611-22. Epub 20140814. PubMed PMID: 25118602; PMCID: PMC4230700. [CrossRef]
  9. Bakhshandeh S, Werner C, Fratzl P, Cipitria A. Microenvironment-mediated cancer dormancy: Insights from metastability theory. Proc Natl Acad Sci U S A. 2022;119(1). PubMed PMID: 34949715; PMCID: PMC8740765. [CrossRef]
  10. Hosseini H, Obradovic MMS, Hoffmann M, Harper KL, Sosa MS, Werner-Klein M, Nanduri LK, Werno C, Ehrl C, Maneck M, Patwary N, Haunschild G, Guzvic M, Reimelt C, Grauvogl M, Eichner N, Weber F, Hartkopf AD, Taran FA, Brucker SY, Fehm T, Rack B, Buchholz S, Spang R, Meister G, Aguirre-Ghiso JA, Klein CA. Early dissemination seeds metastasis in breast cancer. Nature. 2016;540(7634):552-8. Epub 20161214. PubMed PMID: 27974799; PMCID: PMC5390864. [CrossRef]
  11. Cristiano S, Leal A, Phallen J, Fiksel J, Adleff V, Bruhm DC, Jensen SO, Medina JE, Hruban C, White JR, Palsgrove DN, Niknafs N, Anagnostou V, Forde P, Naidoo J, Marrone K, Brahmer J, Woodward BD, Husain H, van Rooijen KL, Orntoft MW, Madsen AH, van de Velde CJH, Verheij M, Cats A, Punt CJA, Vink GR, van Grieken NCT, Koopman M, Fijneman RJA, Johansen JS, Nielsen HJ, Meijer GA, Andersen CL, Scharpf RB, Velculescu VE. Genome-wide cell-free DNA fragmentation in patients with cancer. Nature. 2019;570(7761):385-9. Epub 20190529. PubMed PMID: 31142840; PMCID: PMC6774252. [CrossRef]
  12. Paez-Ribes M, Allen E, Hudock J, Takeda T, Okuyama H, Vinals F, Inoue M, Bergers G, Hanahan D, Casanovas O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 2009;15(3):220-31. PubMed PMID: 19249680; PMCID: PMC2874829. [CrossRef]
  13. Hotz B, Arndt M, Dullat S, Bhargava S, Buhr HJ, Hotz HG. Epithelial to mesenchymal transition: expression of the regulators snail, slug, and twist in pancreatic cancer. Clin Cancer Res. 2007;13(16):4769-76. PubMed PMID: 17699854. [CrossRef]
  14. Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. Loss of E-cadherin promotes metastasis via multiple downstream transcriptional pathways. Cancer Res. 2008;68(10):3645-54. PubMed PMID: 18483246. [CrossRef]
  15. Ozdemir BC, Pentcheva-Hoang T, Carstens JL, Zheng X, Wu CC, Simpson TR, Laklai H, Sugimoto H, Kahlert C, Novitskiy SV, De Jesus-Acosta A, Sharma P, Heidari P, Mahmood U, Chin L, Moses HL, Weaver VM, Maitra A, Allison JP, LeBleu VS, Kalluri R. Depletion of Carcinoma-Associated Fibroblasts and Fibrosis Induces Immunosuppression and Accelerates Pancreas Cancer with Reduced Survival. Cancer Cell. 2015;28(6):831-3. Epub 20151214. PubMed PMID: 28843279. [CrossRef]
  16. Come C, Magnino F, Bibeau F, De Santa Barbara P, Becker KF, Theillet C, Savagner P. Snail and slug play distinct roles during breast carcinoma progression. Clin Cancer Res. 2006;12(18):5395-402. PubMed PMID: 17000672. [CrossRef]
  17. Guo W, Keckesova Z, Donaher JL, Shibue T, Tischler V, Reinhardt F, Itzkovitz S, Noske A, Zurrer-Hardi U, Bell G, Tam WL, Mani SA, van Oudenaarden A, Weinberg RA. Slug and Sox9 cooperatively determine the mammary stem cell state. Cell. 2012;148(5):1015-28. PubMed PMID: 22385965; PMCID: PMC3305806. [CrossRef]
  18. Shih JY, Yang PC. The EMT regulator slug and lung carcinogenesis. Carcinogenesis. 2011;32(9):1299-304. Epub 20110610. PubMed PMID: 21665887. [CrossRef]
  19. Katoh M, Katoh M. Comparative genomics on SNAI1, SNAI2, and SNAI3 orthologs. Oncol Rep. 2005;14(4):1083-6. PubMed PMID: 16142376.
  20. Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA, Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA. Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis. Cell. 2004;117(7):927-39. PubMed PMID: 15210113. [CrossRef]
  21. Eckert MA, Lwin TM, Chang AT, Kim J, Danis E, Ohno-Machado L, Yang J. Twist1-induced invadopodia formation promotes tumor metastasis. Cancer Cell. 2011;19(3):372-86. PubMed PMID: 21397860; PMCID: PMC3072410. [CrossRef]
  22. Wang S, Zhong L, Li Y, Xiao D, Zhang R, Liao D, Lv D, Wang X, Wang J, Xie X, Chen J, Wu Y, Kang T. Up-regulation of PCOLCE by TWIST1 promotes metastasis in Osteosarcoma. Theranostics. 2019;9(15):4342-53. Epub 20190609. PubMed PMID: 31285765; PMCID: PMC6599655. [CrossRef]
  23. Fang X, Cai Y, Liu J, Wang Z, Wu Q, Zhang Z, Yang CJ, Yuan L, Ouyang G. Twist2 contributes to breast cancer progression by promoting an epithelial-mesenchymal transition and cancer stem-like cell self-renewal. Oncogene. 2011;30(47):4707-20. Epub 20110523. PubMed PMID: 21602879. [CrossRef]
  24. Gasparotto D, Polesel J, Marzotto A, Colladel R, Piccinin S, Modena P, Grizzo A, Sulfaro S, Serraino D, Barzan L, Doglioni C, Maestro R. Overexpression of TWIST2 correlates with poor prognosis in head and neck squamous cell carcinomas. Oncotarget. 2011;2(12):1165-75. PubMed PMID: 22201613; PMCID: PMC3282075. [CrossRef]
  25. Safe S, Abdelrahim M. Sp transcription factor family and its role in cancer. Eur J Cancer. 2005;41(16):2438-48. Epub 20051004. PubMed PMID: 16209919. [CrossRef]
  26. Abdelrahim M, Smith R, 3rd, Burghardt R, Safe S. Role of Sp proteins in regulation of vascular endothelial growth factor expression and proliferation of pancreatic cancer cells. Cancer Res. 2004;64(18):6740-9. PubMed PMID: 15374992. [CrossRef]
  27. Beishline K, Azizkhan-Clifford J. Sp1 and the ‘hallmarks of cancer’. FEBS J. 2015;282(2):224-58. Epub 20150108. PubMed PMID: 25393971. [CrossRef]
  28. Sun X, Xiao C, Wang X, Wu S, Yang Z, Sui B, Song Y. Role of post-translational modifications of Sp1 in cancer: state of the art. Front Cell Dev Biol. 2024;12:1412461. Epub 20240820. PubMed PMID: 39228402; PMCID: PMC11368732. [CrossRef]
  29. Deniaud E, Baguet J, Mathieu AL, Pages G, Marvel J, Leverrier Y. Overexpression of Sp1 transcription factor induces apoptosis. Oncogene. 2006;25(53):7096-105. Epub 20060522. PubMed PMID: 16715126. [CrossRef]
  30. Kim KH, Roberts CW. Targeting EZH2 in cancer. Nat Med. 2016;22(2):128-34. PubMed PMID: 26845405; PMCID: PMC4918227. [CrossRef]
  31. Zheng M, Jiang YP, Chen W, Li KD, Liu X, Gao SY, Feng H, Wang SS, Jiang J, Ma XR, Cen X, Tang YJ, Chen Y, Lin YF, Tang YL, Liang XH. Snail and Slug collaborate on EMT and tumor metastasis through miR-101-mediated EZH2 axis in oral tongue squamous cell carcinoma. Oncotarget. 2015;6(9):6797-810. PubMed PMID: 25762643; PMCID: PMC4466650. [CrossRef]
  32. Varambally S, Dhanasekaran SM, Zhou M, Barrette TR, Kumar-Sinha C, Sanda MG, Ghosh D, Pienta KJ, Sewalt RG, Otte AP, Rubin MA, Chinnaiyan AM. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 2002;419(6907):624-9. PubMed PMID: 12374981. [CrossRef]
  33. Chang CJ, Yang JY, Xia W, Chen CT, Xie X, Chao CH, Woodward WA, Hsu JM, Hortobagyi GN, Hung MC. EZH2 promotes expansion of breast tumor initiating cells through activation of RAF1-beta-catenin signaling. Cancer Cell. 2011;19(1):86-100. Epub 20110106. PubMed PMID: 21215703; PMCID: PMC3041516. [CrossRef]
  34. Gong S, Wu C, Duan Y, Tang J, Wu P. A Comprehensive Pan-Cancer Analysis for Pituitary Tumor-Transforming Gene 1. Front Genet. 2022;13:843579. Epub 20220225. PubMed PMID: 35281830; PMCID: PMC8916819. [CrossRef]
  35. Altieri DC. Survivin, cancer networks and pathway-directed drug discovery. Nat Rev Cancer. 2008;8(1):61-70. PubMed PMID: 18075512. [CrossRef]
  36. Lasham A, Print CG, Woolley AG, Dunn SE, Braithwaite AW. YB-1: oncoprotein, prognostic marker and therapeutic target? Biochem J. 2013;449(1):11-23. PubMed PMID: 23216250. [CrossRef]
  37. Johnson JL, Pillai S, Pernazza D, Sebti SM, Lawrence NJ, Chellappan SP. Regulation of matrix metalloproteinase genes by E2F transcription factors: Rb-Raf-1 interaction as a novel target for metastatic disease. Cancer Res. 2012;72(2):516-26. Epub 20111115. PubMed PMID: 22086850; PMCID: PMC3261351. [CrossRef]
  38. Ramsay RG, Gonda TJ. MYB function in normal and cancer cells. Nat Rev Cancer. 2008;8(7):523-34. PubMed PMID: 18574464. [CrossRef]
  39. Kim DJ. The Role of the DNA Methyltransferase Family and the Therapeutic Potential of DNMT Inhibitors in Tumor Treatment. Curr Oncol. 2025;32(2). Epub 20250205. PubMed PMID: 39996888; PMCID: PMC11854558. [CrossRef]
  40. Casalino L, Verde P. Multifaceted Roles of DNA Methylation in Neoplastic Transformation, from Tumor Suppressors to EMT and Metastasis. Genes (Basel). 2020;11(8). Epub 20200812. PubMed PMID: 32806509; PMCID: PMC7463745. [CrossRef]
  41. Wozniak M, Czyz M. Exploring oncogenic roles and clinical significance of EZH2: focus on non-canonical activities. Ther Adv Med Oncol. 2025;17:17588359241306026. Epub 20250107. PubMed PMID: 39776536; PMCID: PMC11705335. [CrossRef]
  42. Liu S, Ye D, Guo W, Yu W, He Y, Hu J, Wang Y, Zhang L, Liao Y, Song H, Zhong S, Xu D, Yin H, Sun B, Wang X, Liu J, Wu Y, Zhou BP, Zhang Z, Deng J. G9a is essential for EMT-mediated metastasis and maintenance of cancer stem cell-like characters in head and neck squamous cell carcinoma. Oncotarget. 2015;6(9):6887-901. PubMed PMID: 25749385; PMCID: PMC4466657. [CrossRef]
  43. Tan T, Shi P, Abbas MN, Wang Y, Xu J, Chen Y, Cui H. Epigenetic modification regulates tumor progression and metastasis through EMT (Review). Int J Oncol. 2022;60(6). Epub 20220421. PubMed PMID: 35445731; PMCID: PMC9084613. [CrossRef]
  44. Iyer NG, Ozdag H, Caldas C. p300/CBP and cancer. Oncogene. 2004;23(24):4225-31. PubMed PMID: 15156177. [CrossRef]
  45. Weichert W. HDAC expression and clinical prognosis in human malignancies. Cancer Lett. 2009;280(2):168-76. Epub 20081221. PubMed PMID: 19103471. [CrossRef]
  46. Kroesen M, Gielen P, Brok IC, Armandari I, Hoogerbrugge PM, Adema GJ. HDAC inhibitors and immunotherapy; a double edged sword? Oncotarget. 2014;5(16):6558-72. PubMed PMID: 25115382; PMCID: PMC4196144. [CrossRef]
  47. Alver BH, Kim KH, Lu P, Wang X, Manchester HE, Wang W, Haswell JR, Park PJ, Roberts CW. The SWI/SNF chromatin remodelling complex is required for maintenance of lineage specific enhancers. Nat Commun. 2017;8:14648. Epub 20170306. PubMed PMID: 28262751; PMCID: PMC5343482. [CrossRef]
  48. Gupta RA, Shah N, Wang KC, Kim J, Horlings HM, Wong DJ, Tsai MC, Hung T, Argani P, Rinn JL, Wang Y, Brzoska P, Kong B, Li R, West RB, van de Vijver MJ, Sukumar S, Chang HY. Long non-coding RNA HOTAIR reprograms chromatin state to promote cancer metastasis. Nature. 2010;464(7291):1071-6. PubMed PMID: 20393566; PMCID: PMC3049919. [CrossRef]
  49. Italiano A, Soria JC, Toulmonde M, Michot JM, Lucchesi C, Varga A, Coindre JM, Blakemore SJ, Clawson A, Suttle B, McDonald AA, Woodruff M, Ribich S, Hedrick E, Keilhack H, Thomson B, Owa T, Copeland RA, Ho PTC, Ribrag V. Tazemetostat, an EZH2 inhibitor, in relapsed or refractory B-cell non-Hodgkin lymphoma and advanced solid tumours: a first-in-human, open-label, phase 1 study. Lancet Oncol. 2018;19(5):649-59. Epub 20180409. PubMed PMID: 29650362. [CrossRef]
  50. Chen IC, Sethy B, Liou JP. Recent Update of HDAC Inhibitors in Lymphoma. Front Cell Dev Biol. 2020;8:576391. Epub 20200903. PubMed PMID: 33015069; PMCID: PMC7494784. [CrossRef]
  51. Alba-Castellon L, Olivera-Salguero R, Mestre-Farrera A, Pena R, Herrera M, Bonilla F, Casal JI, Baulida J, Pena C, Garcia de Herreros A. Snail1-Dependent Activation of Cancer-Associated Fibroblast Controls Epithelial Tumor Cell Invasion and Metastasis. Cancer Res. 2016;76(21):6205-17. Epub 20160808. PubMed PMID: 27503928. [CrossRef]
  52. Wang X, Cui P, Deng Y, Zhang B, Gao Z, Li T, Yin Y, Li J. Function of SP1 in tumors and focused treatment approaches for immune evasion (Review). Oncol Lett. 2025;30(4):483. Epub 20250814. PubMed PMID: 40861097; PMCID: PMC12373422. [CrossRef]
  53. Gao Y, Gan K, Liu K, Xu B, Chen M. SP1 Expression and the Clinicopathological Features of Tumors: A Meta-Analysis and Bioinformatics Analysis. Pathol Oncol Res. 2021;27:581998. Epub 20210128. PubMed PMID: 34257529; PMCID: PMC8262197. [CrossRef]
  54. Wu X, Tao P, Zhou Q, Li J, Yu Z, Wang X, Li J, Li C, Yan M, Zhu Z, Liu B, Su L. IL-6 secreted by cancer-associated fibroblasts promotes epithelial-mesenchymal transition and metastasis of gastric cancer via JAK2/STAT3 signaling pathway. Oncotarget. 2017;8(13):20741-50. PubMed PMID: 28186964; PMCID: PMC5400541. [CrossRef]
  55. Huang B, Lang X, Li X. The role of IL-6/JAK2/STAT3 signaling pathway in cancers. Front Oncol. 2022;12:1023177. Epub 20221216. PubMed PMID: 36591515; PMCID: PMC9800921. [CrossRef]
  56. Liu Q, Li A, Tian Y, Wu JD, Liu Y, Li T, Chen Y, Han X, Wu K. The CXCL8-CXCR1/2 pathways in cancer. Cytokine Growth Factor Rev. 2016;31:61-71. Epub 20160825. PubMed PMID: 27578214; PMCID: PMC6142815. [CrossRef]
  57. Xiong X, Liao X, Qiu S, Xu H, Zhang S, Wang S, Ai J, Yang L. CXCL8 in Tumor Biology and Its Implications for Clinical Translation. Front Mol Biosci. 2022;9:723846. Epub 20220315. PubMed PMID: 35372515; PMCID: PMC8965068. [CrossRef]
  58. Yan M, Zheng M, Niu R, Yang X, Tian S, Fan L, Li Y, Zhang S. Roles of tumor-associated neutrophils in tumor metastasis and its clinical applications. Front Cell Dev Biol. 2022;10:938289. Epub 20220817. PubMed PMID: 36060811; PMCID: PMC9428510. [CrossRef]
  59. Zhou C, Gao Y, Ding P, Wu T, Ji G. The role of CXCL family members in different diseases. Cell Death Discov. 2023;9(1):212. Epub 20230701. PubMed PMID: 37393391; PMCID: PMC10314943. [CrossRef]
  60. Domanska UM, Kruizinga RC, Nagengast WB, Timmer-Bosscha H, Huls G, de Vries EG, Walenkamp AM. A review on CXCR4/CXCL12 axis in oncology: no place to hide. Eur J Cancer. 2013;49(1):219-30. Epub 20120609. PubMed PMID: 22683307. [CrossRef]
  61. Chatterjee S, Behnam Azad B, Nimmagadda S. The intricate role of CXCR4 in cancer. Adv Cancer Res. 2014;124:31-82. PubMed PMID: 25287686; PMCID: PMC4322894. [CrossRef]
  62. Augoff K, Hryniewicz-Jankowska A, Tabola R, Stach K. MMP9: A Tough Target for Targeted Therapy for Cancer. Cancers (Basel). 2022;14(7). Epub 20220406. PubMed PMID: 35406619; PMCID: PMC8998077. [CrossRef]
  63. Quintero-Fabian S, Arreola R, Becerril-Villanueva E, Torres-Romero JC, Arana-Argaez V, Lara-Riegos J, Ramirez-Camacho MA, Alvarez-Sanchez ME. Role of Matrix Metalloproteinases in Angiogenesis and Cancer. Front Oncol. 2019;9:1370. Epub 20191206. PubMed PMID: 31921634; PMCID: PMC6915110. [CrossRef]
  64. Gonzalez-Avila G, Sommer B, Mendoza-Posada DA, Ramos C, Garcia-Hernandez AA, Falfan-Valencia R. Matrix metalloproteinases participation in the metastatic process and their diagnostic and therapeutic applications in cancer. Crit Rev Oncol Hematol. 2019;137:57-83. Epub 20190303. PubMed PMID: 31014516. [CrossRef]
  65. Foley CJ, Luo C, O’Callaghan K, Hinds PW, Covic L, Kuliopulos A. Matrix metalloprotease-1a promotes tumorigenesis and metastasis. J Biol Chem. 2012;287(29):24330-8. Epub 20120509. PubMed PMID: 22573325; PMCID: PMC3397859. [CrossRef]
  66. Xu M, Xu C, Wang R, Tang Q, Zhou Q, Wu W, Wan X, Mo H, Pan J, Wang S. Treating human cancer by targeting EZH2. Genes Dis. 2025;12(3):101313. Epub 20240425. PubMed PMID: 40028035; PMCID: PMC11870178. [CrossRef]
  67. Mortezaee K. CXCL12/CXCR4 axis in the microenvironment of solid tumors: A critical mediator of metastasis. Life Sci. 2020;249:117534. Epub 20200307. PubMed PMID: 32156548. [CrossRef]
  68. Raychaudhuri P, Park HJ. FoxM1: a master regulator of tumor metastasis. Cancer Res. 2011;71(13):4329-33. Epub 20110628. PubMed PMID: 21712406; PMCID: PMC3129416. [CrossRef]
  69. Liao GB, Li XZ, Zeng S, Liu C, Yang SM, Yang L, Hu CJ, Bai JY. Regulation of the master regulator FOXM1 in cancer. Cell Commun Signal. 2018;16(1):57. Epub 20180912. PubMed PMID: 30208972; PMCID: PMC6134757. [CrossRef]
  70. Huang Y, Hong W, Wei X. The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 2022;15(1):129. Epub 20220908. PubMed PMID: 36076302; PMCID: PMC9461252. [CrossRef]
  71. Anand S, Vikramdeo KS, Sudan SK, Sharma A, Acharya S, Khan MA, Singh S, Singh AP. From modulation of cellular plasticity to potentiation of therapeutic resistance: new and emerging roles of MYB transcription factors in human malignancies. Cancer Metastasis Rev. 2024;43(1):409-21. Epub 20231110. PubMed PMID: 37950087; PMCID: PMC11015973. [CrossRef]
  72. Yoon CH, Kim MJ, Lee H, Kim RK, Lim EJ, Yoo KC, Lee GH, Cui YH, Oh YS, Gye MC, Lee YY, Park IC, An S, Hwang SG, Park MJ, Suh Y, Lee SJ. PTTG1 oncogene promotes tumor malignancy via epithelial to mesenchymal transition and expansion of cancer stem cell population. J Biol Chem. 2012;287(23):19516-27. Epub 20120416. PubMed PMID: 22511756; PMCID: PMC3365988. [CrossRef]
  73. Liu X, Zeng W, Zheng D, Tang M, Zhou W. Clinical significance of securin expression in solid cancers: A PRISMA-compliant meta-analysis of published studies and bioinformatics analysis based on TCGA dataset. Medicine (Baltimore). 2022;101(37):e30440. PubMed PMID: 36123907; PMCID: PMC9478268. [CrossRef]
  74. Alkrekshi A, Wang W, Rana PS, Markovic V, Sossey-Alaoui K. A comprehensive review of the functions of YB-1 in cancer stemness, metastasis and drug resistance. Cell Signal. 2021;85:110073. Epub 20210703. PubMed PMID: 34224843; PMCID: PMC8878385. [CrossRef]
  75. D’Costa NM, Lowerison MR, Raven PA, Tan Z, Roberts ME, Shrestha R, Urban MW, Monjaras-Avila CU, Oo HZ, Hurtado-Coll A, Chavez-Munoz C, So AI. Y-box binding protein-1 is crucial in acquired drug resistance development in metastatic clear-cell renal cell carcinoma. J Exp Clin Cancer Res. 2020;39(1):33. Epub 20200210. PubMed PMID: 32041631; PMCID: PMC7011538. [CrossRef]
  76. Faldt Beding A, Larsson P, Helou K, Einbeigi Z, Parris TZ. Pan-cancer analysis identifies BIRC5 as a prognostic biomarker. BMC Cancer. 2022;22(1):322. Epub 20220325. PubMed PMID: 35331169; PMCID: PMC8953143. [CrossRef]
  77. Siragusa G, Tomasello L, Giordano C, Pizzolanti G. Survivin (BIRC5): Implications in cancer therapy. Life Sci. 2024;350:122788. Epub 20240605. PubMed PMID: 38848940. [CrossRef]
  78. Lin J, Zhang P, Liu W, Liu G, Zhang J, Yan M, Duan Y, Yang N. A positive feedback loop between ZEB2 and ACSL4 regulates lipid metabolism to promote breast cancer metastasis. Elife. 2023;12. Epub 20231211. PubMed PMID: 38078907; PMCID: PMC10712958. [CrossRef]
  79. Parfenyev SE, Daks AA, Shuvalov OY, Fedorova OA, Pestov NB, Korneenko TV, Barlev NA. Dualistic role of ZEB1 and ZEB2 in tumor progression. Biol Direct. 2025;20(1):32. Epub 20250320. PubMed PMID: 40114235; PMCID: PMC11927373. [CrossRef]
  80. McCormack VA, dos Santos Silva I. Breast density and parenchymal patterns as markers of breast cancer risk: a meta-analysis. Cancer Epidemiol Biomarkers Prev. 2006;15(6):1159-69. PubMed PMID: 16775176. [CrossRef]
  81. Hebert JD, Myers SA, Naba A, Abbruzzese G, Lamar JM, Carr SA, Hynes RO. Proteomic Profiling of the ECM of Xenograft Breast Cancer Metastases in Different Organs Reveals Distinct Metastatic Niches. Cancer Res. 2020;80(7):1475-85. Epub 20200204. PubMed PMID: 32019869; PMCID: PMC7127975. [CrossRef]
  82. Fang S, Dai Y, Mei Y, Yang M, Hu L, Yang H, Guan X, Li J. Clinical significance and biological role of cancer-derived Type I collagen in lung and esophageal cancers. Thorac Cancer. 2019;10(2):277-88. Epub 20190103. PubMed PMID: 30604926; PMCID: PMC6360244. [CrossRef]
  83. Fogg KC, Renner CM, Christian H, Walker A, Marty-Santos L, Khan A, Olson WR, Parent C, O’Shea A, Wellik DM, Weisman PS, Kreeger PK. Ovarian Cells Have Increased Proliferation in Response to Heparin-Binding Epidermal Growth Factor as Collagen Density Increases. Tissue Eng Part A. 2020;26(13-14):747-58. Epub 20200625. PubMed PMID: 32598229; PMCID: PMC7398436. [CrossRef]
  84. van ‘t Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT, Schreiber GJ, Kerkhoven RM, Roberts C, Linsley PS, Bernards R, Friend SH. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002;415(6871):530-6. PubMed PMID: 11823860. [CrossRef]
  85. Lokeshwar VB, Mirza S, Jordan A. Targeting hyaluronic acid family for cancer chemoprevention and therapy. Adv Cancer Res. 2014;123:35-65. PubMed PMID: 25081525; PMCID: PMC4791948. [CrossRef]
  86. Bae YK, Kim A, Kim MK, Choi JE, Kang SH, Lee SJ. Fibronectin expression in carcinoma cells correlates with tumor aggressiveness and poor clinical outcome in patients with invasive breast cancer. Hum Pathol. 2013;44(10):2028-37. Epub 20130514. PubMed PMID: 23684510. [CrossRef]
  87. Gudjonsson T, Ronnov-Jessen L, Villadsen R, Rank F, Bissell MJ, Petersen OW. Normal and tumor-derived myoepithelial cells differ in their ability to interact with luminal breast epithelial cells for polarity and basement membrane deposition. J Cell Sci. 2002;115(Pt 1):39-50. PubMed PMID: 11801722; PMCID: PMC2933194. [CrossRef]
  88. Guess CM, Quaranta V. Defining the role of laminin-332 in carcinoma. Matrix Biol. 2009;28(8):445-55. Epub 20090815. PubMed PMID: 19686849; PMCID: PMC2875997. [CrossRef]
  89. Pearce OMT, Delaine-Smith RM, Maniati E, Nichols S, Wang J, Böhm S, Rajeeve V, Ullah D, Chakravarty P, Jones RR, Montfort A, Dowe T, Gribben J, Jones JL, Kocher HM, Serody JS, Vincent BG, Connelly J, Brenton JD, Chelala C, Cutillas PR, Lockley M, Bessant C, Knight MM, Balkwill FR. Deconstruction of a Metastatic Tumor Microenvironment Reveals a Common Matrix Response in Human Cancers. Cancer Discovery. 2018;8(3):304-19. PubMed PMID: WOS:000426997500021. [CrossRef]
  90. Gocheva V, Naba A, Bhutkar A, Guardia T, Miller KM, Li CM, Dayton TL, Sanchez-Rivera FJ, Kim-Kiselak C, Jailkhani N, Winslow MM, Del Rosario A, Hynes RO, Jacks T. Quantitative proteomics identify Tenascin-C as a promoter of lung cancer progression and contributor to a signature prognostic of patient survival. Proc Natl Acad Sci U S A. 2017;114(28):E5625-E34. Epub 20170626. PubMed PMID: 28652369; PMCID: PMC5514763. [CrossRef]
  91. Xiao W, Wang S, Zhang R, Sohrabi A, Yu Q, Liu S, Ehsanipour A, Liang J, Bierman RD, Nathanson DA, Seidlits SK. Bioengineered scaffolds for 3D culture demonstrate extracellular matrix-mediated mechanisms of chemotherapy resistance in glioblastoma. Matrix Biol. 2020;85-86:128-46. Epub 20190424. PubMed PMID: 31028838; PMCID: PMC6813884. [CrossRef]
  92. Vihinen P, Ala-aho R, Kahari VM. Matrix metalloproteinases as therapeutic targets in cancer. Curr Cancer Drug Targets. 2005;5(3):203-20. PubMed PMID: 15892620. [CrossRef]
  93. Overall CM, Kleifeld O. Tumour microenvironment - Opinion - Validating matrix metalloproteinases as drug targets and anti-targets for cancer therapy. Nature Reviews Cancer. 2006;6(3):227-39. PubMed PMID: WOS:000235591900017. [CrossRef]
  94. Bergers G, Brekken R, McMahon G, Vu TH, Itoh T, Tamaki K, Tanzawa K, Thorpe P, Itohara S, Werb Z, Hanahan D. Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat Cell Biol. 2000;2(10):737-44. PubMed PMID: 11025665; PMCID: PMC2852586. [CrossRef]
  95. Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582-98. PubMed PMID: 27550820. [CrossRef]
  96. Duffy MJ, Duggan C. The urokinase plasminogen activator system: a rich source of tumour markers for the individualised management of patients with cancer. Clin Biochem. 2004;37(7):541-8. PubMed PMID: 15234235. [CrossRef]
  97. Gocheva V, Joyce JA. Cysteine cathepsins and the cutting edge of cancer invasion. Cell Cycle. 2007;6(1):60-4. Epub 20070106. PubMed PMID: 17245112. [CrossRef]
  98. Erler JT, Bennewith KL, Nicolau M, Dornhofer N, Kong C, Le QT, Chi JT, Jeffrey SS, Giaccia AJ. Lysyl oxidase is essential for hypoxia-induced metastasis. Nature. 2006;440(7088):1222-6. PubMed PMID: 16642001. [CrossRef]
  99. Peinado H, Del Carmen Iglesias-de la Cruz M, Olmeda D, Csiszar K, Fong KS, Vega S, Nieto MA, Cano A, Portillo F. A molecular role for lysyl oxidase-like 2 enzyme in snail regulation and tumor progression. EMBO J. 2005;24(19):3446-58. Epub 20050818. PubMed PMID: 16096638; PMCID: PMC1276164. [CrossRef]
  100. Vlodavsky I, Singh P, Boyango I, Gutter-Kapon L, Elkin M, Sanderson RD, Ilan N. Heparanase: From basic research to therapeutic applications in cancer and inflammation. Drug Resist Updat. 2016;29:54-75. Epub 20161006. PubMed PMID: 27912844; PMCID: PMC5447241. [CrossRef]
  101. Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, Vonderheide RH, Pittet MJ, Jain RK, Zou W, Howcroft TK, Woodhouse EC, Weinberg RA, Krummel MF. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24(5):541-50. Epub 20180423. PubMed PMID: 29686425; PMCID: PMC5998822. [CrossRef]
  102. Schumacher D, Strilic B, Sivaraj KK, Wettschureck N, Offermanns S. Platelet-derived nucleotides promote tumor-cell transendothelial migration and metastasis via P2Y2 receptor. Cancer Cell. 2013;24(1):130-7. Epub 20130627. PubMed PMID: 23810565. [CrossRef]
  103. Berghoff AS, Venur VA, Preusser M, Ahluwalia MS. Immune Checkpoint Inhibitors in Brain Metastases: From Biology to Treatment. Am Soc Clin Oncol Educ Book. 2016;35:e116-22. PubMed PMID: 27249713. [CrossRef]
  104. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21(3):418-29. PubMed PMID: 22439937; PMCID: PMC3371414. [CrossRef]
  105. Muller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, McClanahan T, Murphy E, Yuan W, Wagner SN, Barrera JL, Mohar A, Verastegui E, Zlotnik A. Involvement of chemokine receptors in breast cancer metastasis. Nature. 2001;410(6824):50-6. PubMed PMID: 11242036. [CrossRef]
  106. Cabioglu N, Yazici MS, Arun B, Broglio KR, Hortobagyi GN, Price JE, Sahin A. CCR7 and CXCR4 as novel biomarkers predicting axillary lymph node metastasis in T1 breast cancer. Clin Cancer Res. 2005;11(16):5686-93. PubMed PMID: 16115904. [CrossRef]
  107. Xu B, Deng C, Wu X, Ji T, Zhao L, Han Y, Yang W, Qi Y, Wang Z, Yang Z, Yang Y. CCR9 and CCL25: A review of their roles in tumor promotion. J Cell Physiol. 2020;235(12):9121-32. Epub 20200513. PubMed PMID: 32401349. [CrossRef]
  108. Mitsui E, Kikuchi S, Okura T, Tazawa H, Une Y, Nishiwaki N, Kuroda S, Noma K, Kagawa S, Ohara T, Ohtsuka J, Ohki R, Fujiwara T. Novel treatment strategy targeting interleukin-6 induced by cancer associated fibroblasts for peritoneal metastasis of gastric cancer. Sci Rep. 2025;15(1):3267. Epub 20250125. PubMed PMID: 39863722; PMCID: PMC11762302. [CrossRef]
  109. Han YP, Tuan TL, Wu H, Hughes M, Garner WL. TNF-alpha stimulates activation of pro-MMP2 in human skin through NF-(kappa)B mediated induction of MT1-MMP. J Cell Sci. 2001;114(Pt 1):131-9. PubMed PMID: 11112697; PMCID: PMC2435089. [CrossRef]
  110. Colak S, Ten Dijke P. Targeting TGF-beta Signaling in Cancer. Trends Cancer. 2017;3(1):56-71. Epub 20170103. PubMed PMID: 28718426. [CrossRef]
  111. Waugh DJ, Wilson C. The interleukin-8 pathway in cancer. Clin Cancer Res. 2008;14(21):6735-41. PubMed PMID: 18980965. [CrossRef]
  112. Eisenblaetter M, Flores-Borja F, Lee JJ, Wefers C, Smith H, Hueting R, Cooper MS, Blower PJ, Patel D, Rodriguez-Justo M, Milewicz H, Vogl T, Roth J, Tutt A, Schaeffter T, Ng T. Visualization of Tumor-Immune Interaction - Target-Specific Imaging of S100A8/A9 Reveals Pre-Metastatic Niche Establishment. Theranostics. 2017;7(9):2392-401. PubMed PMID: WOS:000403704900004. [CrossRef]
  113. Steeg PS. Targeting metastasis. Nat Rev Cancer. 2016;16(4):201-18. PubMed PMID: 27009393; PMCID: PMC7055530. [CrossRef]
  114. Kaplan RN, Riba RD, Zacharoulis S, Bramley AH, Vincent L, Costa C, MacDonald DD, Jin DK, Shido K, Kerns SA, Zhu Z, Hicklin D, Wu Y, Port JL, Altorki N, Port ER, Ruggero D, Shmelkov SV, Jensen KK, Rafii S, Lyden D. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature. 2005;438(7069):820-7. PubMed PMID: 16341007; PMCID: PMC2945882. [CrossRef]
  115. Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, Le QT, Giaccia AJ. Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 2009;15(1):35-44. PubMed PMID: 19111879; PMCID: PMC3050620. [CrossRef]
  116. Peinado H, Zhang H, Matei IR, Costa-Silva B, Hoshino A, Rodrigues G, Psaila B, Kaplan RN, Bromberg JF, Kang Y, Bissell MJ, Cox TR, Giaccia AJ, Erler JT, Hiratsuka S, Ghajar CM, Lyden D. Pre-metastatic niches: organ-specific homes for metastases. Nat Rev Cancer. 2017;17(5):302-17. Epub 20170317. PubMed PMID: 28303905. [CrossRef]
  117. Grant G, Ferrer CM. The role of the immune tumor microenvironment in shaping metastatic dissemination, dormancy, and outgrowth. Trends Cell Biol. 2025. Epub 20250704. PubMed PMID: 40628544. [CrossRef]
  118. Endo H, Okuyama H, Ohue M, Inoue M. Dormancy of cancer cells with suppression of AKT activity contributes to survival in chronic hypoxia. PLoS One. 2014;9(6):e98858. Epub 20140606. PubMed PMID: 24905002; PMCID: PMC4048292. [CrossRef]
  119. Pouyssegur J, Dayan F, Mazure NM. Hypoxia signalling in cancer and approaches to enforce tumour regression. Nature. 2006;441(7092):437-43. PubMed PMID: 16724055. [CrossRef]
  120. Inoue M, Hager JH, Ferrara N, Gerber HP, Hanahan D. VEGF-A has a critical, nonredundant role in angiogenic switching and pancreatic beta cell carcinogenesis. Cancer Cell. 2002;1(2):193-202. PubMed PMID: 12086877. [CrossRef]
  121. Takeda T, Okuyama H, Nishizawa Y, Tomita S, Inoue M. Hypoxia inducible factor-1alpha is necessary for invasive phenotype in Vegf-deleted islet cell tumors. Sci Rep. 2012;2:494. Epub 20120704. PubMed PMID: 22768384; PMCID: PMC3389366. [CrossRef]
  122. Harada H, Inoue M, Itasaka S, Hirota K, Morinibu A, Shinomiya K, Zeng L, Ou G, Zhu Y, Yoshimura M, McKenna WG, Muschel RJ, Hiraoka M. Cancer cells that survive radiation therapy acquire HIF-1 activity and translocate towards tumour blood vessels. Nat Commun. 2012;3:783. Epub 20120417. PubMed PMID: 22510688; PMCID: PMC3337987. [CrossRef]
  123. Qannita RA, Alalami AI, Harb AA, Aleidi SM, Taneera J, Abu-Gharbieh E, El-Huneidi W, Saleh MA, Alzoubi KH, Semreen MH, Hudaib M, Bustanji Y. Targeting Hypoxia-Inducible Factor-1 (HIF-1) in Cancer: Emerging Therapeutic Strategies and Pathway Regulation. Pharmaceuticals (Basel). 2024;17(2). Epub 20240201. PubMed PMID: 38399410; PMCID: PMC10892333. [CrossRef]
  124. Ameri K, Jahangiri A, Rajah AM, Tormos KV, Nagarajan R, Pekmezci M, Nguyen V, Wheeler ML, Murphy MP, Sanders TA, Jeffrey SS, Yeghiazarians Y, Rinaudo PF, Costello JF, Aghi MK, Maltepe E. HIGD1A Regulates Oxygen Consumption, ROS Production, and AMPK Activity during Glucose Deprivation to Modulate Cell Survival and Tumor Growth. Cell Rep. 2015;10(6):891-9. Epub 20150213. PubMed PMID: 25683712; PMCID: PMC4534363. [CrossRef]
  125. Aguirre-Ghiso JA, Estrada Y, Liu D, Ossowski L. ERK(MAPK) activity as a determinant of tumor growth and dormancy; regulation by p38(SAPK). Cancer Res. 2003;63(7):1684-95. PubMed PMID: 12670923.
  126. Bui T, Gu Y, Ancot F, Sanguin-Gendreau V, Zuo D, Muller WJ. Emergence of beta1 integrin-deficient breast tumours from dormancy involves both inactivation of p53 and generation of a permissive tumour microenvironment. Oncogene. 2022;41(4):527-37. Epub 20211115. PubMed PMID: 34782719; PMCID: PMC8782722. [CrossRef]
  127. Nam JM, Onodera Y, Bissell MJ, Park CC. Breast cancer cells in three-dimensional culture display an enhanced radioresponse after coordinate targeting of integrin alpha5beta1 and fibronectin. Cancer Res. 2010;70(13):5238-48. Epub 20100601. PubMed PMID: 20516121; PMCID: PMC2933183. [CrossRef]
  128. Huang T, Sun L, Yuan X, Qiu H. Thrombospondin-1 is a multifaceted player in tumor progression. Oncotarget. 2017;8(48):84546-58. Epub 20170711. PubMed PMID: 29137447; PMCID: PMC5663619. [CrossRef]
  129. Choi YH, Burdick MD, Strieter BA, Mehrad B, Strieter RM. CXCR4, but not CXCR7, discriminates metastatic behavior in non-small cell lung cancer cells. Mol Cancer Res. 2014;12(1):38-47. Epub 20130911. PubMed PMID: 24025971; PMCID: PMC4262747. [CrossRef]
  130. Sosa MS, Parikh F, Maia AG, Estrada Y, Bosch A, Bragado P, Ekpin E, George A, Zheng Y, Lam HM, Morrissey C, Chung CY, Farias EF, Bernstein E, Aguirre-Ghiso JA. NR2F1 controls tumour cell dormancy via SOX9- and RARbeta-driven quiescence programmes. Nat Commun. 2015;6:6170. Epub 20150130. PubMed PMID: 25636082; PMCID: PMC4313575. [CrossRef]
  131. Barnieh FM, Morton J, Olanrewaju O, El-Khamisy SF. Decoding the adaptive survival mechanisms of breast cancer dormancy. Oncogene. 2025;44(40):3759-73. Epub 20250827. PubMed PMID: 40866498; PMCID: PMC12477053. [CrossRef]
  132. Park SL, Buzzai A, Rautela J, Hor JL, Hochheiser K, Effern M, McBain N, Wagner T, Edwards J, McConville R, Wilmott JS, Scolyer RA, Tuting T, Palendira U, Gyorki D, Mueller SN, Huntington ND, Bedoui S, Holzel M, Mackay LK, Waithman J, Gebhardt T. Tissue-resident memory CD8(+) T cells promote melanoma-immune equilibrium in skin. Nature. 2019;565(7739):366-71. Epub 20181231. PubMed PMID: 30598548. [CrossRef]
  133. Karacz CM, Yan J, Zhu H, Gerber DE. Timing, Sites, and Correlates of Lung Cancer Recurrence. Clin Lung Cancer. 2020;21(2):127-35 e3. Epub 20191220. PubMed PMID: 31932216; PMCID: PMC7061059. [CrossRef]
  134. Shin JH, Yoo HB, Roe JS. Current advances and future directions in targeting histone demethylases for cancer therapy. Mol Cells. 2025;48(3):100192. Epub 20250210. PubMed PMID: 39938867; PMCID: PMC11889978. [CrossRef]
  135. Xu Y, Li N, Xiang R, Sun P. Emerging roles of the p38 MAPK and PI3K/AKT/mTOR pathways in oncogene-induced senescence. Trends Biochem Sci. 2014;39(6):268-76. Epub 20140509. PubMed PMID: 24818748; PMCID: PMC4358807. [CrossRef]
  136. Di Martino JS, Nobre AR, Mondal C, Taha I, Farias EF, Fertig EJ, Naba A, Aguirre-Ghiso JA, Bravo-Cordero JJ. A tumor-derived type III collagen-rich ECM niche regulates tumor cell dormancy. Nat Cancer. 2022;3(1):90-107. Epub 20211213. PubMed PMID: 35121989; PMCID: PMC8818089. [CrossRef]
  137. Fox DB, Garcia NMG, McKinney BJ, Lupo R, Noteware LC, Newcomb R, Liu J, Locasale JW, Hirschey MD, Alvarez JV. NRF2 activation promotes the recurrence of dormant tumour cells through regulation of redox and nucleotide metabolism. Nat Metab. 2020;2(4):318-34. Epub 20200420. PubMed PMID: 32691018; PMCID: PMC7370851. [CrossRef]
  138. Kaur A, Webster MR, Marchbank K, Behera R, Ndoye A, Kugel CH, 3rd, Dang VM, Appleton J, O’Connell MP, Cheng P, Valiga AA, Morissette R, McDonnell NB, Ferrucci L, Kossenkov AV, Meeth K, Tang HY, Yin X, Wood WH, 3rd, Lehrmann E, Becker KG, Flaherty KT, Frederick DT, Wargo JA, Cooper ZA, Tetzlaff MT, Hudgens C, Aird KM, Zhang R, Xu X, Liu Q, Bartlett E, Karakousis G, Eroglu Z, Lo RS, Chan M, Menzies AM, Long GV, Johnson DB, Sosman J, Schilling B, Schadendorf D, Speicher DW, Bosenberg M, Ribas A, Weeraratna AT. sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature. 2016;532(7598):250-4. Epub 20160404. PubMed PMID: 27042933; PMCID: PMC4833579. [CrossRef]
  139. Prunier C, Baker D, Ten Dijke P, Ritsma L. TGF-beta Family Signaling Pathways in Cellular Dormancy. Trends Cancer. 2019;5(1):66-78. Epub 20181128. PubMed PMID: 30616757. [CrossRef]
  140. Kurppa KJ, Liu Y, To C, Zhang T, Fan M, Vajdi A, Knelson EH, Xie Y, Lim K, Cejas P, Portell A, Lizotte PH, Ficarro SB, Li S, Chen T, Haikala HM, Wang H, Bahcall M, Gao Y, Shalhout S, Boettcher S, Shin BH, Thai T, Wilkens MK, Tillgren ML, Mushajiang M, Xu M, Choi J, Bertram AA, Ebert BL, Beroukhim R, Bandopadhayay P, Awad MM, Gokhale PC, Kirschmeier PT, Marto JA, Camargo FD, Haq R, Paweletz CP, Wong KK, Barbie DA, Long HW, Gray NS, Janne PA. Treatment-Induced Tumor Dormancy through YAP-Mediated Transcriptional Reprogramming of the Apoptotic Pathway. Cancer Cell. 2020;37(1):104-22 e12. PubMed PMID: 31935369; PMCID: PMC7146079. [CrossRef]
  141. Lotter W, Hassett MJ, Schultz N, Kehl KL, Van Allen EM, Cerami E. Artificial Intelligence in Oncology: Current Landscape, Challenges, and Future Directions. Cancer Discov. 2024;14(5):711-26. PubMed PMID: 38597966; PMCID: PMC11131133. [CrossRef]
  142. Vandekerkhove G, Lavoie JM, Annala M, Murtha AJ, Sundahl N, Walz S, Sano T, Taavitsainen S, Ritch E, Fazli L, Hurtado-Coll A, Wang G, Nykter M, Black PC, Todenhofer T, Ost P, Gibb EA, Chi KN, Eigl BJ, Wyatt AW. Plasma ctDNA is a tumor tissue surrogate and enables clinical-genomic stratification of metastatic bladder cancer. Nat Commun. 2021;12(1):184. Epub 20210108. PubMed PMID: 33420073; PMCID: PMC7794518. [CrossRef]
  143. Liu B, Tang L, Peng N, Wang L. Lung and bone metastases patterns in limb osteosarcoma: Surgical treatment of primary site improves overall survival. Medicine (Baltimore). 2023;102(42):e35671. PubMed PMID: 37861481; PMCID: PMC10589517. [CrossRef]
  144. Silva JAM, Marchiori E, Amorim VB, Barreto MM. CT features of osteosarcoma lung metastasis: a retrospective study of 127 patients. J Bras Pneumol. 2023;49(2):e20220433. Epub 20230428. PubMed PMID: 37132704; PMCID: PMC10171270. [CrossRef]
  145. Marko TA, Diessner BJ, Spector LG. Prevalence of Metastasis at Diagnosis of Osteosarcoma: An International Comparison. Pediatr Blood Cancer. 2016;63(6):1006-11. Epub 20160229. PubMed PMID: 26929018; PMCID: PMC4833631. [CrossRef]
  146. Harris MA, Hawkins CJ. Recent and Ongoing Research into Metastatic Osteosarcoma Treatments. Int J Mol Sci. 2022;23(7). Epub 20220330. PubMed PMID: 35409176; PMCID: PMC8998815. [CrossRef]
  147. Jiang Y, Wang J, Sun M, Zuo D, Wang H, Shen J, Jiang W, Mu H, Ma X, Yin F, Lin J, Wang C, Yu S, Jiang L, Lv G, Liu F, Xue L, Tian K, Wang G, Zhou Z, Lv Y, Wang Z, Zhang T, Xu J, Yang L, Zhao K, Sun W, Tang Y, Cai Z, Wang S, Hua Y. Multi-omics analysis identifies osteosarcoma subtypes with distinct prognosis indicating stratified treatment. Nat Commun. 2022;13(1):7207. Epub 20221123. PubMed PMID: 36418292; PMCID: PMC9684515. [CrossRef]
  148. Beird HC, Wu CC, Nakazawa M, Ingram D, Daniele JR, Lazcano R, Little L, Davies C, Daw NC, Wani K, Wang WL, Song X, Gumbs C, Zhang J, Rubin B, Conley A, Flanagan AM, Lazar AJ, Futreal PA. Complete loss of TP53 and RB1 is associated with complex genome and low immune infiltrate in pleomorphic rhabdomyosarcoma. HGG Adv. 2023;4(4):100224. Epub 20230719. PubMed PMID: 37593416; PMCID: PMC10428123. [CrossRef]
  149. Zhong L, Liao D, Li J, Liu W, Wang J, Zeng C, Wang X, Cao Z, Zhang R, Li M, Jiang K, Zeng YX, Sui J, Kang T. Rab22a-NeoF1 fusion protein promotes osteosarcoma lung metastasis through its secretion into exosomes. Signal Transduct Target Ther. 2021;6(1):59. Epub 20210211. PubMed PMID: 33568623; PMCID: PMC7876000. [CrossRef]
  150. Jiang Y, Li F, Gao B, Ma M, Chen M, Wu Y, Zhang W, Sun Y, Liu S, Shen H. KDM6B-mediated histone demethylation of LDHA promotes lung metastasis of osteosarcoma. Theranostics. 2021;11(8):3868-81. Epub 20210206. PubMed PMID: 33664867; PMCID: PMC7914357. [CrossRef]
  151. Zhang Y, Xu Y, Bao Y, Luo Y, Qiu G, He M, Lu J, Xu J, Chen B, Wang Y. N6-methyladenosine (m6A) modification in osteosarcoma: expression, function and interaction with noncoding RNAs - an updated review. Epigenetics. 2023;18(1):2260213. Epub 20230928. PubMed PMID: 37766615; PMCID: PMC10540650. [CrossRef]
  152. Feng Z, Ou Y, Hao L. The roles of glycolysis in osteosarcoma. Front Pharmacol. 2022;13:950886. Epub 20220817. PubMed PMID: 36059961; PMCID: PMC9428632. [CrossRef]
  153. Luo ZW, Liu PP, Wang ZX, Chen CY, Xie H. Macrophages in Osteosarcoma Immune Microenvironment: Implications for Immunotherapy. Front Oncol. 2020;10:586580. Epub 20201210. PubMed PMID: 33363016; PMCID: PMC7758531. [CrossRef]
  154. Zhou J, Liu T, Wang W. Prognostic significance of matrix metalloproteinase 9 expression in osteosarcoma: A meta-analysis of 16 studies. Medicine (Baltimore). 2018;97(44):e13051. PubMed PMID: 30383677; PMCID: PMC6221749. [CrossRef]
  155. Lucotti S, Muschel RJ. Platelets and Metastasis: New Implications of an Old Interplay. Front Oncol. 2020;10:1350. Epub 20200918. PubMed PMID: 33042789; PMCID: PMC7530207. [CrossRef]
  156. McAloney CA, Makkawi R, Budhathoki Y, Cannon MV, Franz EM, Gross AC, Cam M, Vetter TA, Duhen R, Davies AE, Roberts RD. Host-derived growth factors drive ERK phosphorylation and MCL1 expression to promote osteosarcoma cell survival during metastatic lung colonization. Cell Oncol (Dordr). 2024;47(1):259-82. Epub 20230907. PubMed PMID: 37676378; PMCID: PMC10899530. [CrossRef]
  157. Du X, Wei H, Zhang B, Wang B, Li Z, Pang LK, Zhao R, Yao W. Molecular mechanisms of osteosarcoma metastasis and possible treatment opportunities. Front Oncol. 2023;13:1117867. Epub 20230501. PubMed PMID: 37197432; PMCID: PMC10183593. [CrossRef]
  158. Li B, Wang Z, Wu H, Xue M, Lin P, Wang S, Lin N, Huang X, Pan W, Liu M, Yan X, Qu H, Sun L, Li H, Wu Y, Teng W, Wang Z, Zhou X, Chen H, Poznansky MC, Ye Z. Epigenetic Regulation of CXCL12 Plays a Critical Role in Mediating Tumor Progression and the Immune Response In Osteosarcoma. Cancer Res. 2018;78(14):3938-53. Epub 20180507. PubMed PMID: 29735547. [CrossRef]
  159. Gvozdenovic A, Boro A, Meier D, Bode-Lesniewska B, Born W, Muff R, Fuchs B. Targeting alphavbeta3 and alphavbeta5 integrins inhibits pulmonary metastasis in an intratibial xenograft osteosarcoma mouse model. Oncotarget. 2016;7(34):55141-54. PubMed PMID: 27409827; PMCID: PMC5342407. [CrossRef]
  160. Grisez BT, Ray JJ, Bostian PA, Markel JE, Lindsey BA. Highly metastatic K7M2 cell line: A novel murine model capable of in vivo imaging via luciferase vector transfection. J Orthop Res. 2018. Epub 20180210. PubMed PMID: 29427436; PMCID: PMC6086764. [CrossRef]
  161. Berman SD, Calo E, Landman AS, Danielian PS, Miller ES, West JC, Fonhoue BD, Caron A, Bronson R, Bouxsein ML, Mukherjee S, Lees JA. Metastatic osteosarcoma induced by inactivation of Rb and p53 in the osteoblast lineage. Proc Natl Acad Sci U S A. 2008;105(33):11851-6. Epub 20080812. PubMed PMID: 18697945; PMCID: PMC2575280. [CrossRef]
  162. Lamhamedi-Cherradi SE, Mohiuddin S, Mishra DK, Krishnan S, Velasco AR, Vetter AM, Pence K, McCall D, Truong DD, Cuglievan B, Menegaz BA, Utama B, Daw NC, Molina ER, Zielinski RJ, Livingston JA, Gorlick R, Mikos AG, Kim MP, Ludwig JA. Transcriptional activators YAP/TAZ and AXL orchestrate dedifferentiation, cell fate, and metastasis in human osteosarcoma. Cancer Gene Ther. 2021;28(12):1325-38. Epub 20210106. PubMed PMID: 33408328; PMCID: PMC8636268. [CrossRef]
  163. Just MA, Van Mater D, Wagner LM. Receptor tyrosine kinase inhibitors for the treatment of osteosarcoma and Ewing sarcoma. Pediatr Blood Cancer. 2021;68(8):e29084. Epub 20210424. PubMed PMID: 33894051; PMCID: PMC8238849. [CrossRef]
  164. Bhalla S, Gerber DE. AXL Inhibitors: Status of Clinical Development. Curr Oncol Rep. 2023;25(5):521-9. Epub 20230315. PubMed PMID: 36920638; PMCID: PMC11161200. [CrossRef]
  165. Nirala BK, Yamamichi T, Yustein JT. Deciphering the Signaling Mechanisms of Osteosarcoma Tumorigenesis. Int J Mol Sci. 2023;24(14). Epub 20230712. PubMed PMID: 37511127; PMCID: PMC10379831. [CrossRef]
  166. Yu X, Yustein JT, Xu J. Research models and mesenchymal/epithelial plasticity of osteosarcoma. Cell Biosci. 2021;11(1):94. Epub 20210522. PubMed PMID: 34022967; PMCID: PMC8141200. [CrossRef]
  167. Imamura T. Physiological functions and underlying mechanisms of fibroblast growth factor (FGF) family members: recent findings and implications for their pharmacological application. Biol Pharm Bull. 2014;37(7):1081-9. PubMed PMID: 24988999. [CrossRef]
  168. Sbaraglia M, Bellan E, Dei Tos AP. The 2020 WHO Classification of Soft Tissue Tumours: news and perspectives. Pathologica. 2021;113(2):70-84. Epub 20201103. PubMed PMID: 33179614; PMCID: PMC8167394. [CrossRef]
  169. Bovee JV, Hogendoorn PC, Wunder JS, Alman BA. Cartilage tumours and bone development: molecular pathology and possible therapeutic targets. Nat Rev Cancer. 2010;10(7):481-8. Epub 20100610. PubMed PMID: 20535132. [CrossRef]
  170. Amary MF, Bacsi K, Maggiani F, Damato S, Halai D, Berisha F, Pollock R, O’Donnell P, Grigoriadis A, Diss T, Eskandarpour M, Presneau N, Hogendoorn PC, Futreal A, Tirabosco R, Flanagan AM. IDH1 and IDH2 mutations are frequent events in central chondrosarcoma and central and periosteal chondromas but not in other mesenchymal tumours. J Pathol. 2011;224(3):334-43. Epub 20110519. PubMed PMID: 21598255. [CrossRef]
  171. Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, Marks KM, Prins RM, Ward PS, Yen KE, Liau LM, Rabinowitz JD, Cantley LC, Thompson CB, Vander Heiden MG, Su SM. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2010;465(7300):966. PubMed PMID: 20559394; PMCID: PMC3766976. [CrossRef]
  172. Evans HL, Ayala AG, Romsdahl MM. Prognostic factors in chondrosarcoma of bone: a clinicopathologic analysis with emphasis on histologic grading. Cancer. 1977;40(2):818-31. PubMed PMID: 890662. [CrossRef]
  173. Ingangi V, De Chiara A, Ferrara G, Gallo M, Catapano A, Fazioli F, Di Carluccio G, Peranzoni E, Marigo I, Carriero MV, Minopoli M. Emerging Treatments Targeting the Tumor Microenvironment for Advanced Chondrosarcoma. Cells. 2024;13(11). Epub 20240604. PubMed PMID: 38891109; PMCID: PMC11171855. [CrossRef]
  174. Syrjanen KJ. Spontaneous evolution of intraepithelial lesions according to the grade and type of the implicated human papillomavirus (HPV). Eur J Obstet Gynecol Reprod Biol. 1996;65(1):45-53. PubMed PMID: 8706956. [CrossRef]
  175. A S, Kani V, Vasudevan S, Esakki M. Dedifferentiated Chondrosarcoma: A Report of a Rare and Intriguing Case. Cureus. 2024;16(9):e68452. Epub 20240902. PubMed PMID: 39360119; PMCID: PMC11446496. [CrossRef]
  176. Yang J, Lou S, Yao T. Trends in primary malignant bone cancer incidence and mortality in the United States, 2000-2017: A population-based study. J Bone Oncol. 2024;46:100607. Epub 20240511. PubMed PMID: 38778835; PMCID: PMC11109025. [CrossRef]
  177. van Praag Veroniek VM, Rueten-Budde AJ, Ho V, Dijkstra PDS, Study group B, Soft tissue t, Fiocco M, van de Sande MAJ. Incidence, outcomes and prognostic factors during 25 years of treatment of chondrosarcomas. Surg Oncol. 2018;27(3):402-8. Epub 20180506. PubMed PMID: 30217294. [CrossRef]
  178. Nicolle R, Ayadi M, Gomez-Brouchet A, Armenoult L, Banneau G, Elarouci N, Tallegas M, Decouvelaere AV, Aubert S, Redini F, Marie B, Labit-Bouvier C, Reina N, Karanian M, le Nail LR, Anract P, Gouin F, Larousserie F, de Reynies A, de Pinieux G. Integrated molecular characterization of chondrosarcoma reveals critical determinants of disease progression. Nat Commun. 2019;10(1):4622. Epub 20191011. PubMed PMID: 31604924; PMCID: PMC6789144. [CrossRef]
  179. Campbell VT, Nadesan P, Ali SA, Wang CY, Whetstone H, Poon R, Wei Q, Keilty J, Proctor J, Wang LW, Apte SS, McGovern K, Alman BA, Wunder JS. Hedgehog pathway inhibition in chondrosarcoma using the smoothened inhibitor IPI-926 directly inhibits sarcoma cell growth. Mol Cancer Ther. 2014;13(5):1259-69. Epub 20140314. PubMed PMID: 24634412. [CrossRef]
  180. Walker RL, Hornicek FJ, Duan Z. Advances in the Molecular Biology of Chondrosarcoma for Drug Discovery and Precision Medicine. Cancers (Basel). 2025;17(16). Epub 20250819. PubMed PMID: 40867318; PMCID: PMC12385095. [CrossRef]
  181. Iozzo RV, Sanderson RD. Proteoglycans in cancer biology, tumour microenvironment and angiogenesis. J Cell Mol Med. 2011;15(5):1013-31. PubMed PMID: 21155971; PMCID: PMC3633488. [CrossRef]
  182. Cui N, Hu M, Khalil RA. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog Mol Biol Transl Sci. 2017;147:1-73. Epub 20170322. PubMed PMID: 28413025; PMCID: PMC5430303. [CrossRef]
  183. Lau YK, Gobin AM, West JL. Overexpression of lysyl oxidase to increase matrix crosslinking and improve tissue strength in dermal wound healing. Ann Biomed Eng. 2006;34(8):1239-46. Epub 20060628. PubMed PMID: 16804742. [CrossRef]
  184. Levental KR, Yu H, Kass L, Lakins JN, Egeblad M, Erler JT, Fong SF, Csiszar K, Giaccia A, Weninger W, Yamauchi M, Gasser DL, Weaver VM. Matrix crosslinking forces tumor progression by enhancing integrin signaling. Cell. 2009;139(5):891-906. PubMed PMID: 19931152; PMCID: PMC2788004. [CrossRef]
  185. Semenza GL. Hypoxia-inducible factors in physiology and medicine. Cell. 2012;148(3):399-408. PubMed PMID: 22304911; PMCID: PMC3437543. [CrossRef]
  186. Hou SM, Lin CY, Fong YC, Tang CH. Hypoxia-regulated exosomes mediate M2 macrophage polarization and promote metastasis in chondrosarcoma. Aging (Albany NY). 2023;15(22):13163-75. Epub 20231121. PubMed PMID: 37993261; PMCID: PMC10713415. [CrossRef]
  187. Cammarota F, Laukkanen MO. Mesenchymal Stem/Stromal Cells in Stromal Evolution and Cancer Progression. Stem Cells Int. 2016;2016:4824573. Epub 20151221. PubMed PMID: 26798356; PMCID: PMC4699086. [CrossRef]
  188. Zhang L, Zhou F, ten Dijke P. Signaling interplay between transforming growth factor-beta receptor and PI3K/AKT pathways in cancer. Trends Biochem Sci. 2013;38(12):612-20. Epub 20131114. PubMed PMID: 24239264. [CrossRef]
  189. Meijer DM, Ruano D, Briaire-de Bruijn IH, Wijers-Koster PM, van de Sande MAJ, Gelderblom H, Cleton-Jansen AM, de Miranda N, Kuijjer ML, Bovee J. The Variable Genomic Landscape During Osteosarcoma Progression: Insights From a Longitudinal WGS Analysis. Genes Chromosomes Cancer. 2024;63(7):e23253. PubMed PMID: 39023390. [CrossRef]
  190. Wada T, Nakashima T, Hiroshi N, Penninger JM. RANKL-RANK signaling in osteoclastogenesis and bone disease. Trends Mol Med. 2006;12(1):17-25. Epub 20051213. PubMed PMID: 16356770. [CrossRef]
  191. Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3(10):721-32. PubMed PMID: 13130303. [CrossRef]
  192. Jones RL, Katz D, Loggers ET, Davidson D, Rodler ET, Pollack SM. Clinical benefit of antiangiogenic therapy in advanced and metastatic chondrosarcoma. Med Oncol. 2017;34(10):167. Epub 20170829. PubMed PMID: 28852958; PMCID: PMC5574947. [CrossRef]
  193. Su Z, Ho JWK, Yau RCH, Lam YL, Shek TWH, Yeung MCF, Chen H, Oreffo ROC, Cheah KSE, Cheung KSC. A single-cell atlas of conventional central chondrosarcoma reveals the role of endoplasmic reticulum stress in malignant transformation. Commun Biol. 2024;7(1):124. Epub 20240124. PubMed PMID: 38267611; PMCID: PMC10808239. [CrossRef]
  194. Dermawan JKT, Nafa K, Mohanty A, Xu Y, Rijo I, Casanova J, Villafania L, Benhamida J, Kelly CM, Tap WD, Boland PJ, Fabbri N, Healey JH, Ladanyi M, Lu C, Hameed M. Distinct IDH1/2-associated Methylation Profile and Enrichment of TP53 and TERT Mutations Distinguish Dedifferentiated Chondrosarcoma from Conventional Chondrosarcoma. Cancer Res Commun. 2023;3(3):431-43. Epub 20230314. PubMed PMID: 36926116; PMCID: PMC10013202. [CrossRef]
  195. Lacuna KP, Ingham M, Chen L, Das B, Lee SM, Ge L, Druta M, Conley AP, Keohan ML, Agulnik M, Burgess MA, Chalmers AW, D’Amato GZ, Powers B, Seetharam M, Siontis BL, Pelosof LC, Van Tine BA, Schwartz GK, Weiss MC. Correlative results from NCI CTEP/ETCTN 10330: A phase 2 study of belinostat with SGI-110 (guadecitabine) or ASTX727 (decitabine/cedazuridine) for advanced conventional chondrosarcoma (cCS). J Clin Oncol. 2024;42(16). PubMed PMID: WOS:001275557402684.
  196. Sheikh TN, Chen X, Xu X, McGuire JT, Ingham M, Lu C, Schwartz GK. Growth Inhibition and Induction of Innate Immune Signaling of Chondrosarcomas with Epigenetic Inhibitors. Mol Cancer Ther. 2021;20(12):2362-71. Epub 20210922. PubMed PMID: 34552007; PMCID: PMC8643315. [CrossRef]
  197. Dai W, Qiao X, Fang Y, Guo R, Bai P, Liu S, Li T, Jiang Y, Wei S, Na Z, Xiao X, Li D. Epigenetics-targeted drugs: current paradigms and future challenges. Signal Transduct Target Ther. 2024;9(1):332. Epub 20241126. PubMed PMID: 39592582; PMCID: PMC11627502. [CrossRef]
  198. Oza J, Lee SM, Weiss MC, Siontis BL, Powers BC, Chow WA, Magana W, Sheikh T, Piekarz R, Schwartz GK, Ingham M. A phase 2 study of belinostat and SGI-110 (guadecitabine) for the treatment of unresectable and metastatic conventional chondrosarcoma. J Clin Oncol. 2021;39(15). PubMed PMID: WOS:000708120306169. [CrossRef]
  199. Micaily I, Roche M, Ibrahim MY, Martinez-Outschoorn U, Mallick AB. Metabolic Pathways and Targets in Chondrosarcoma. Front Oncol. 2021;11:772263. Epub 20211206. PubMed PMID: 34938658; PMCID: PMC8685273. [CrossRef]
  200. Bansal A, Goyal S, Goyal A, Jana M. WHO classification of soft tissue tumours 2020: An update and simplified approach for radiologists. Eur J Radiol. 2021;143:109937. Epub 20210828. PubMed PMID: 34547634. [CrossRef]
  201. Schmitz E, Nessim C. Retroperitoneal Sarcoma Care in 2021. Cancers (Basel). 2022;14(5). Epub 20220302. PubMed PMID: 35267600; PMCID: PMC8909774. [CrossRef]
  202. Engstrom K, Willen H, Kabjorn-Gustafsson C, Andersson C, Olsson M, Goransson M, Jarnum S, Olofsson A, Warnhammar E, Aman P. The myxoid/round cell liposarcoma fusion oncogene FUS-DDIT3 and the normal DDIT3 induce a liposarcoma phenotype in transfected human fibrosarcoma cells. Am J Pathol. 2006;168(5):1642-53. PubMed PMID: 16651630; PMCID: PMC1606602. [CrossRef]
  203. Wang L, Ren W, Zhou X, Sheng W, Wang J. Pleomorphic liposarcoma: a clinicopathological, immunohistochemical and molecular cytogenetic study of 32 additional cases. Pathol Int. 2013;63(11):523-31. PubMed PMID: 24274714. [CrossRef]
  204. Jonczak E, Grossman J, Alessandrino F, Seldon Taswell C, Velez-Torres JM, Trent J. Liposarcoma: A Journey into a Rare Tumor’s Epidemiology, Diagnosis, Pathophysiology, and Limitations of Current Therapies. Cancers (Basel). 2024;16(22). Epub 20241118. PubMed PMID: 39594813; PMCID: PMC11592651. [CrossRef]
  205. Haddox CL, Hornick JL, Roland CL, Baldini EH, Keedy VL, Riedel RF. Diagnosis and management of dedifferentiated liposarcoma: A multidisciplinary position statement. Cancer Treat Rev. 2024;131:102846. Epub 20241018. PubMed PMID: 39454547. [CrossRef]
  206. Mujtaba B, Wang F, Taher A, Aslam R, Madewell JE, Nassar S. Myxoid Liposarcoma With Skeletal Metastases: Pathophysiology and Imaging Characteristics. Curr Probl Diagn Radiol. 2021;50(1):66-73. Epub 20191030. PubMed PMID: 31813645. [CrossRef]
  207. Wan L, Tu C, Qi L, Li Z. Survivorship and prognostic factors for pleomorphic liposarcoma: a population-based study. J Orthop Surg Res. 2021;16(1):175. Epub 20210304. PubMed PMID: 33663547; PMCID: PMC7931523. [CrossRef]
  208. James J, Yajid AI, Yahaya S, Abdullah S, Sharif SET. Prognostic Implications of and Co-amplification in Liposarcoma: Insights from FISH analysis for Translational Oncology. Clin Transl Oncol. 2025. PubMed PMID: WOS:001529127500001. [CrossRef]
  209. Hou X, Shi W, Luo W, Luo Y, Huang X, Li J, Ji N, Chen Q. FUS::DDIT3 Fusion Protein in the Development of Myxoid Liposarcoma and Possible Implications for Therapy. Biomolecules. 2024;14(10). Epub 20241014. PubMed PMID: 39456230; PMCID: PMC11506083. [CrossRef]
  210. Gruel N, Quignot C, Lesage L, El Zein S, Bonvalot S, Tzanis D, Ait Rais K, Quinquis F, Manciot B, Vibert J, El Tannir N, Dahmani A, Derrien H, Decaudin D, Bieche I, Courtois L, Mariani O, Linares LK, Gayte L, Baulande S, Waterfall JJ, Delattre O, Pierron G, Watson S. Cellular origin and clonal evolution of human dedifferentiated liposarcoma. Nat Commun. 2024;15(1):7941. Epub 20240912. PubMed PMID: 39266532; PMCID: PMC11393420. [CrossRef]
  211. Benesova I, Kalkusova K, Kwon YS, Taborska P, Stakheev D, Krausova K, Smetanova J, Ozaniak A, Bartunkova J, Smrz D, Strizova ZO. Cancer-associated fibroblasts in human malignancies, with a particular emphasis on sarcomas (Review). Int J Oncol. 2025;67(4). Epub 20250808. PubMed PMID: 40776758; PMCID: PMC12370362. [CrossRef]
  212. Vautrot V, Hervieu A, Bertaut A, Charon-Barra C, Naiken I, Causseret S, Chaigneau L, Desmoulins I, Rederstoff E, Isambert N, Gobbo J. Small Extracellular Vesicles as Biomarkers in Sarcoma Follow-Up: Protocol for a Prospective, Multicentric Pilot Study. JMIR Res Protoc. 2025;14:e63718. Epub 20250909. PubMed PMID: 40925003; PMCID: PMC12457855. [CrossRef]
  213. Torres MB, Leung CH, Zoghbi M, Lazcano R, Ingram D, Wani K, Keung EZ, Zarzour MA, Scally CP, Hunt KK, Conley A, Bishop AJ, Guadagnolo BA, Farooqi A, Mitra D, Yoder AK, Nakazawa MS, Araujo D, Livingston A, Ratan R, Patel S, Ravi V, Lazar AJ, Roland CL, Somaiah N, Nassif Haddad EF. Dedifferentiated liposarcomas treated with immune checkpoint blockade: the MD Anderson experience. Front Immunol. 2025;16:1567736. Epub 20250430. PubMed PMID: 40370451; PMCID: PMC12075363. [CrossRef]
  214. Schmid A, Eisenhardt AE, Bogner B, Runkel A, Lausch U, Pauli T, Antolini LN, Boneberg A, Kiefer J, Bronsert P, Boerries M, Eisenhardt SU, Braig D. Intratumoral heterogeneity of cancer driver genomic alterations in myxoid liposarcomas. Cancer. 2025;131(12):e35937. PubMed PMID: 40489430; PMCID: PMC12148203. [CrossRef]
  215. Versari I, Salucci S, Bavelloni A, Battistelli M, Traversari M, Wang A, Sampaolesi M, Faenza I. The Emerging Role and Clinical Significance of PI3K-Akt-mTOR in Rhabdomyosarcoma. Biomolecules. 2025;15(3). Epub 20250225. PubMed PMID: 40149870; PMCID: PMC11940244. [CrossRef]
  216. Kruiswijk AA, Kuhrij LS, Dorleijn DMJ, van de Sande MAJ, van Bodegom-Vos L, Marang-van de Mheen PJ. Follow-Up after Curative Surgical Treatment of Soft-Tissue Sarcoma for Early Detection of Recurrence: Which Patients Have More or Fewer Visits than Advised in Guidelines? Cancers (Basel). 2023;15(18). Epub 20230918. PubMed PMID: 37760585; PMCID: PMC10527323. [CrossRef]
  217. Kerrison WGJ, Lee ATJ, Thway K, Jones RL, Huang PH. Current Status and Future Directions of Immunotherapies in Soft Tissue Sarcomas. Biomedicines. 2022;10(3). Epub 20220228. PubMed PMID: 35327375; PMCID: PMC8945421. [CrossRef]
  218. Liu H, Wang X, Wang X, Qiu F, Zhou B. Challenges and hope: latest research trends in the clinical treatment and prognosis of liposarcoma. Front Pharmacol. 2025;16:1529755. Epub 20250512. PubMed PMID: 40421219; PMCID: PMC12104207. [CrossRef]
  219. von Mehren M, Kane JM, Agulnik M, Bui MM, Carr-Ascher J, Choy E, Connelly M, Dry S, Ganjoo KN, Gonzalez RJ, Holder A, Homsi J, Keedy V, Kelly CM, Kim E, Liebner D, McCarter M, McGarry SV, Mesko NW, Meyer C, Pappo AS, Parkes AM, Petersen IA, Pollack SM, Poppe M, Riedel RF, Schuetze S, Shabason J, Sicklick JK, Spraker MB, Zimel M, Hang LE, Sundar H, Bergman MA. Soft Tissue Sarcoma, Version 2.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2022;20(7):815-33. PubMed PMID: 35830886; PMCID: PMC10186762. [CrossRef]
  220. Resag A, Toffanin G, Benesova I, Muller L, Potkrajcic V, Ozaniak A, Lischke R, Bartunkova J, Rosato A, Johrens K, Eckert F, Strizova Z, Schmitz M. The Immune Contexture of Liposarcoma and Its Clinical Implications. Cancers (Basel). 2022;14(19). Epub 20220921. PubMed PMID: 36230502; PMCID: PMC9559230. [CrossRef]
  221. Somaiah N, Tap W. MDM2-p53 in liposarcoma: The need for targeted therapies with novel mechanisms of action. Cancer Treat Rev. 2024;122:102668. Epub 20231210. PubMed PMID: 38104352. [CrossRef]
  222. Ostrowski SM, Fisher DE. Biology of Melanoma. Hematol Oncol Clin North Am. 2021;35(1):29-56. Epub 20201026. PubMed PMID: 33759772. [CrossRef]
  223. Mataca E, Migaldi M, Cesinaro AM. Impact of Dermoscopy and Reflectance Confocal Microscopy on the Histopathologic Diagnosis of Lentigo Maligna/Lentigo Maligna Melanoma. Am J Dermatopathol. 2018;40(12):884-9. PubMed PMID: 29933314. [CrossRef]
  224. Switzer B, Puzanov I, Skitzki JJ, Hamad L, Ernstoff MS. Managing Metastatic Melanoma in 2022: A Clinical Review. JCO Oncol Pract. 2022;18(5):335-51. Epub 20220208. PubMed PMID: 35133862; PMCID: PMC9810138. [CrossRef]
  225. Adler NR, McArthur GA, Mar VJ. Lymphatic and Hematogenous Dissemination in Patients With Primary Cutaneous Melanoma. JAMA Dermatol. 2019;155(11):1322. PubMed PMID: 31509186. [CrossRef]
  226. Caruso G, Garcia Moreira CG, Iaboni E, Tripodo M, Ferrarotto R, Abbritti RV, Conte L, Caffo M. Tumor Microenvironment in Melanoma Brain Metastasis: A New Potential Target? Int J Mol Sci. 2025;26(11). Epub 20250523. PubMed PMID: 40507830; PMCID: PMC12154486. [CrossRef]
  227. Rhodin KE, Fimbres DP, Burner DN, Hollander S, O’Connor MH, Beasley GM. Melanoma lymph node metastases - moving beyond quantity in clinical trial design and contemporary practice. Front Oncol. 2022;12:1021057. Epub 20221014. PubMed PMID: 36411863; PMCID: PMC9675405. [CrossRef]
  228. Wagstaff W, Mwamba RN, Grullon K, Armstrong M, Zhao P, Hendren-Santiago B, Qin KH, Li AJ, Hu DA, Youssef A, Reid RR, Luu HH, Shen L, He TC, Haydon RC. Melanoma: Molecular genetics, metastasis, targeted therapies, immunotherapies, and therapeutic resistance. Genes Dis. 2022;9(6):1608-23. Epub 20220427. PubMed PMID: 36157497; PMCID: PMC9485270. [CrossRef]
  229. Braden J, Conway JW, Wilmott JS, Scolyer RA, Long GV, da Silva IP. Do BRAF-targeted therapies have a role in the era of immunotherapy? ESMO Open. 2025;10(7):105314. Epub 20250620. PubMed PMID: 40543211; PMCID: PMC12221719. [CrossRef]
  230. Kolathur KK, Nag R, Shenoy PV, Malik Y, Varanasi SM, Angom RS, Mukhopadhyay D. Molecular Susceptibility and Treatment Challenges in Melanoma. Cells. 2024;13(16). Epub 20240820. PubMed PMID: 39195270; PMCID: PMC11352263. [CrossRef]
  231. Yang TT, Yu S, Ke CK, Cheng ST. The Genomic Landscape of Melanoma and Its Therapeutic Implications. Genes (Basel). 2023;14(5). Epub 20230429. PubMed PMID: 37239381; PMCID: PMC10218388. [CrossRef]
  232. Guo W, Wang H, Li C. Signal pathways of melanoma and targeted therapy. Signal Transduct Target Ther. 2021;6(1):424. Epub 20211220. PubMed PMID: 34924562; PMCID: PMC8685279. [CrossRef]
  233. Lade-Keller J, Riber-Hansen R, Guldberg P, Schmidt H, Hamilton-Dutoit SJ, Steiniche T. E- to N-cadherin switch in melanoma is associated with decreased expression of phosphatase and tensin homolog and cancer progression. Br J Dermatol. 2013;169(3):618-28. PubMed PMID: 23662813. [CrossRef]
  234. Chen Y, Yan Y, Wei W. Research advances of matrix metalloproteinases family in uveal melanoma. Eur J Med Res. 2025;30(1):609. Epub 20250709. PubMed PMID: 40635067; PMCID: PMC12239450. [CrossRef]
  235. Hunter MV, Joshi E, Bowker S, Montal E, Ma Y, Kim YH, Yang Z, Tuffery L, Li Z, Rosiek E, Browning A, Moncada R, Yanai I, Byrne H, Monetti M, de Stanchina E, Hamard PJ, Koche RP, White RM. Mechanical confinement governs phenotypic plasticity in melanoma. Nature. 2025. Epub 20250827. PubMed PMID: 40866703. [CrossRef]
  236. Reiss Y, Proudfoot AE, Power CA, Campbell JJ, Butcher EC. CC chemokine receptor (CCR)4 and the CCR10 ligand cutaneous T cell-attracting chemokine (CTACK) in lymphocyte trafficking to inflamed skin. J Exp Med. 2001;194(10):1541-7. PubMed PMID: 11714760; PMCID: PMC2193675. [CrossRef]
  237. Sahai E, Astsaturov I, Cukierman E, DeNardo DG, Egeblad M, Evans RM, Fearon D, Greten FR, Hingorani SR, Hunter T, Hynes RO, Jain RK, Janowitz T, Jorgensen C, Kimmelman AC, Kolonin MG, Maki RG, Powers RS, Pure E, Ramirez DC, Scherz-Shouval R, Sherman MH, Stewart S, Tlsty TD, Tuveson DA, Watt FM, Weaver V, Weeraratna AT, Werb Z. A framework for advancing our understanding of cancer-associated fibroblasts. Nat Rev Cancer. 2020;20(3):174-86. Epub 20200124. PubMed PMID: 31980749; PMCID: PMC7046529. [CrossRef]
  238. Basak U, Sarkar T, Mukherjee S, Chakraborty S, Dutta A, Dutta S, Nayak D, Kaushik S, Das T, Sa G. Tumor-associated macrophages: an effective player of the tumor microenvironment. Front Immunol. 2023;14:1295257. Epub 20231116. PubMed PMID: 38035101; PMCID: PMC10687432. [CrossRef]
  239. Sun M, Wu J. Molecular and immune landscape of melanoma: a risk stratification model for precision oncology. Discov Oncol. 2025;16(1):667. Epub 20250504. PubMed PMID: 40319421; PMCID: PMC12050255. [CrossRef]
  240. Ju W, Cai HH, Zheng W, Li DM, Zhang W, Yang XH, Yan ZX. Cross-talk between lymphangiogenesis and malignant melanoma cells: New opinions on tumour drainage and immunization (Review). Oncol Lett. 2024;27(2):81. Epub 20240105. PubMed PMID: 38249813; PMCID: PMC10797314. [CrossRef]
  241. Ahmadi E, Chiari C, Madina BR, Yarovinsky TO, Krady MM, Chen J, Almassian B, Nakaar V, Wang K. CARG-2020 targets IL-12, IL-17, and PD-L1 pathways to effectively treat melanoma and breast cancer. Sci Rep. 2025;15(1):29649. Epub 20250813. PubMed PMID: 40804279; PMCID: PMC12350727. [CrossRef]
  242. Knight A, Karapetyan L, Kirkwood JM. Immunotherapy in Melanoma: Recent Advances and Future Directions. Cancers (Basel). 2023;15(4). Epub 20230209. PubMed PMID: 36831449; PMCID: PMC9954703. [CrossRef]
  243. Michielon E, de Gruijl TD, Gibbs S. From simplicity to complexity in current melanoma models. Exp Dermatol. 2022;31(12):1818-36. Epub 20221005. PubMed PMID: 36103206; PMCID: PMC10092692. [CrossRef]
  244. Hu A, Sun L, Lin H, Liao Y, Yang H, Mao Y. Harnessing innate immune pathways for therapeutic advancement in cancer. Signal Transduct Target Ther. 2024;9(1):68. Epub 20240325. PubMed PMID: 38523155; PMCID: PMC10961329. [CrossRef]
  245. Ernst M, Giubellino A. The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma. Biomedicines. 2022;10(4). Epub 20220331. PubMed PMID: 35453572; PMCID: PMC9029866. [CrossRef]
  246. Reschke R, Enk AH, Hassel JC. Prognostic Biomarkers in Evolving Melanoma Immunotherapy. Am J Clin Dermatol. 2025;26(2):213-23. Epub 20241221. PubMed PMID: 39707058; PMCID: PMC11850490. [CrossRef]
  247. Patton EE, Mueller KL, Adams DJ, Anandasabapathy N, Aplin AE, Bertolotto C, Bosenberg M, Ceol CJ, Burd CE, Chi P, Herlyn M, Holmen SL, Karreth FA, Kaufman CK, Khan S, Kobold S, Leucci E, Levy C, Lombard DB, Lund AW, Marie KL, Marine JC, Marais R, McMahon M, Robles-Espinoza CD, Ronai ZA, Samuels Y, Soengas MS, Villanueva J, Weeraratna AT, White RM, Yeh I, Zhu J, Zon LI, Hurlbert MS, Merlino G. Melanoma models for the next generation of therapies. Cancer Cell. 2021;39(5):610-31. Epub 20210204. PubMed PMID: 33545064; PMCID: PMC8378471. [CrossRef]
  248. Singh SP, Madke T, Chand P. Global Epidemiology of Hepatocellular Carcinoma. J Clin Exp Hepatol. 2025;15(2):102446. Epub 20241028. PubMed PMID: 39659901; PMCID: PMC11626783. [CrossRef]
  249. Gan C, Yuan Y, Shen H, Gao J, Kong X, Che Z, Guo Y, Wang H, Dong E, Xiao J. Liver diseases: epidemiology, causes, trends and predictions. Signal Transduct Target Ther. 2025;10(1):33. Epub 20250205. PubMed PMID: 39904973; PMCID: PMC11794951. [CrossRef]
  250. da Fonseca LG, Araujo RLC. Fibrolamellar hepatocellular carcinoma: Advances, challenges and opportunities in a rare malignancy. World J Gastrointest Surg. 2025;17(10):109107. PubMed PMID: 41178855; PMCID: PMC12576631. [CrossRef]
  251. Llovet JM, Kelley RK, Villanueva A, Singal AG, Pikarsky E, Roayaie S, Lencioni R, Koike K, Zucman-Rossi J, Finn RS. Hepatocellular carcinoma. Nat Rev Dis Primers. 2021;7(1):6. Epub 20210121. PubMed PMID: 33479224. [CrossRef]
  252. Salimiaghdam N, Mustafa A, Pokuaa IO, Hamidi A, Chen E. Recurrent Aggressive Hepatocellular Carcinoma Presenting With Chest Wall Metastasis and Portal Vein Thrombosis: A Rare Case and a Multidisciplinary Perspective. Cureus. 2025;17(7):e87330. Epub 20250705. PubMed PMID: 40762000; PMCID: PMC12320916. [CrossRef]
  253. Macedo F, Ladeira K, Pinho F, Saraiva N, Bonito N, Pinto L, Goncalves F. Bone Metastases: An Overview. Oncol Rev. 2017;11(1):321. Epub 20170509. PubMed PMID: 28584570; PMCID: PMC5444408. [CrossRef]
  254. Terada T, Maruo H. Unusual extrahepatic metastatic sites from hepatocellular carcinoma. Int J Clin Exp Pathol. 2013;6(5):816-20. Epub 20130415. PubMed PMID: 23638212; PMCID: PMC3638091.
  255. Wang S, Wang A, Lin J, Xie Y, Wu L, Huang H, Bian J, Yang X, Wan X, Zhao H, Huang J. Brain metastases from hepatocellular carcinoma: recent advances and future avenues. Oncotarget. 2017;8(15):25814-29. PubMed PMID: 28445959; PMCID: PMC5421971. [CrossRef]
  256. Ramaite FT, Nkadimeng SM. Targeting inflammatory pathways in hepatocellular carcinoma: recent developments. Discov Oncol. 2025;16(1):1174. Epub 20250622. PubMed PMID: 40544399; PMCID: PMC12183140. [CrossRef]
  257. Jang JW, Kim JS, Kim HS, Tak KY, Lee SK, Nam HC, Sung PS, Kim CM, Park JY, Bae SH, Choi JY, Yoon SK. Significance of TERT Genetic Alterations and Telomere Length in Hepatocellular Carcinoma. Cancers (Basel). 2021;13(9). Epub 20210430. PubMed PMID: 33946181; PMCID: PMC8125722. [CrossRef]
  258. Li J, Bai L, Xin ZD, Song JJ, Chen H, Song XB, Zhou J. TERT-TP53 mutations: a novel biomarker pair for hepatocellular carcinoma recurrence and prognosis (vol 15, 3620, 2025). Sci Rep-Uk. 2025;15(1). PubMed PMID: WOS:001518931600015. [CrossRef]
  259. Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. beta-Catenin signaling in hepatocellular carcinoma. J Clin Invest. 2022;132(4). PubMed PMID: 35166233; PMCID: PMC8843739. [CrossRef]
  260. Luo X, He X, Zhang X, Zhao X, Zhang Y, Shi Y, Hua S. Hepatocellular carcinoma: signaling pathways, targeted therapy, and immunotherapy. MedComm (2020). 2024;5(2):e474. Epub 20240204. PubMed PMID: 38318160; PMCID: PMC10838672. [CrossRef]
  261. Arii S, Mise M, Harada T, Furutani M, Ishigami S, Niwano M, Mizumoto M, Fukumoto M, Imamura M. Overexpression of matrix metalloproteinase 9 gene in hepatocellular carcinoma with invasive potential. Hepatology. 1996;24(2):316-22. PubMed PMID: 8690399. [CrossRef]
  262. Benderski K, Schneider P, Kordeves P, Fichter M, Schunke J, De Lorenzi F, Durak F, Schrors B, Akilli O, Kiessling F, Bros M, Diken M, Grabbe S, Schattenberg JM, Lammers T, Sofias AM, Kaps L. A hepatocellular carcinoma model with and without parenchymal liver damage that integrates technical and pathophysiological advantages for therapy testing. Pharmacol Res. 2025;211:107560. Epub 20241225. PubMed PMID: 39730106. [CrossRef]
  263. Masuda H. Cancer-associated fibroblasts in cancer drug resistance and cancer progression: a review. Cell Death Discov. 2025;11(1):341. Epub 20250724. PubMed PMID: 40707476; PMCID: PMC12289933. [CrossRef]
  264. Ge J, Jiang H, Chen J, Chen X, Zhang Y, Shi L, Zheng X, Jiang J, Chen L. TGF-beta signaling orchestrates cancer-associated fibroblasts in the tumor microenvironment of human hepatocellular carcinoma: unveiling insights and clinical significance. BMC Cancer. 2025;25(1):113. Epub 20250121. PubMed PMID: 39838288; PMCID: PMC11753146. [CrossRef]
  265. Bannister ME, Chatterjee DA, Shetty S, Patten DA. The Role of Macrophages in Hepatocellular Carcinoma and Their Therapeutic Potential. Int J Mol Sci. 2024;25(23). Epub 20241207. PubMed PMID: 39684877; PMCID: PMC11641991. [CrossRef]
  266. Jinno K, Tanimizu M, Hyodo I, Nishikawa Y, Hosokawa Y, Doi T, Endo H, Yamashita T, Okada Y. Circulating vascular endothelial growth factor (VEGF) is a possible tumor marker for metastasis in human hepatocellular carcinoma. J Gastroenterol. 1998;33(3):376-82. PubMed PMID: 9658317. [CrossRef]
  267. Kurma K, Manches O, Chuffart F, Sturm N, Gharzeddine K, Zhang J, Mercey-Ressejac M, Rousseaux S, Millet A, Lerat H, Marche PN, Macek Jilkova Z, Decaens T. DEN-Induced Rat Model Reproduces Key Features of Human Hepatocellular Carcinoma. Cancers (Basel). 2021;13(19). Epub 20211004. PubMed PMID: 34638465; PMCID: PMC8508319. [CrossRef]
  268. Stribbling SM, Beach C, Ryan AJ. Orthotopic and metastatic tumour models in preclinical cancer research. Pharmacol Ther. 2024;257:108631. Epub 20240311. PubMed PMID: 38467308; PMCID: PMC11781865. [CrossRef]
  269. Zhang X, Zhang C, Zhang Z, Zhang X. Multidimensional insights into exosomes in hepatocellular carcinoma: from genesis to clinical application. Front Immunol. 2025;16:1628573. Epub 20250813. PubMed PMID: 40881702; PMCID: PMC12380686. [CrossRef]
  270. Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther. 2024;9(1):270. Epub 20241011. PubMed PMID: 39389953; PMCID: PMC11467208. [CrossRef]
  271. Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther. 2025;10(1):35. Epub 20250207. PubMed PMID: 39915447; PMCID: PMC11802921. [CrossRef]
  272. Xu J, Ding L, Mei J, Hu Y, Kong X, Dai S, Bu T, Xiao Q, Ding K. Dual roles and therapeutic targeting of tumor-associated macrophages in tumor microenvironments. Signal Transduct Target Ther. 2025;10(1):268. Epub 20250825. PubMed PMID: 40850976; PMCID: PMC12375796. [CrossRef]
  273. Alrumaihi F. Role of Liquid Biopsy for Early Detection, Prognosis, and Therapeutic Monitoring of Hepatocellular Carcinoma. Diagnostics (Basel). 2025;15(13). Epub 20250628. PubMed PMID: 40647654; PMCID: PMC12249386. [CrossRef]
  274. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, Hawkins C, Ng HK, Pfister SM, Reifenberger G, Soffietti R, von Deimling A, Ellison DW. The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro Oncol. 2021;23(8):1231-51. PubMed PMID: 34185076; PMCID: PMC8328013. [CrossRef]
  275. Hambardzumyan D, Bergers G. Glioblastoma: Defining Tumor Niches. Trends Cancer. 2015;1(4):252-65. PubMed PMID: 27088132; PMCID: PMC4831073. [CrossRef]
  276. Singh S, Dey D, Barik D, Mohapatra I, Kim S, Sharma M, Prasad S, Wang P, Singh A, Singh G. Glioblastoma at the crossroads: current understanding and future therapeutic horizons. Signal Transduct Target Ther. 2025;10(1):213. Epub 20250709. PubMed PMID: 40628732; PMCID: PMC12238593. [CrossRef]
  277. Kurdi M, Baeesa S, Okal F, Bamaga AK, Faizo E, Fathaddin AA, Alkhotani A, Karami MM, Bahakeem B. Extracranial metastasis of brain glioblastoma outside CNS: Pathogenesis revisited. Cancer Rep (Hoboken). 2023;6(12):e1905. Epub 20231009. PubMed PMID: 37814403; PMCID: PMC10728524. [CrossRef]
  278. Iacob G, Dinca EB. Current data and strategy in glioblastoma multiforme. J Med Life. 2009;2(4):386-93. PubMed PMID: 20108752; PMCID: PMC3019011.
  279. Verhaak RG, Hoadley KA, Purdom E, Wang V, Qi Y, Wilkerson MD, Miller CR, Ding L, Golub T, Mesirov JP, Alexe G, Lawrence M, O’Kelly M, Tamayo P, Weir BA, Gabriel S, Winckler W, Gupta S, Jakkula L, Feiler HS, Hodgson JG, James CD, Sarkaria JN, Brennan C, Kahn A, Spellman PT, Wilson RK, Speed TP, Gray JW, Meyerson M, Getz G, Perou CM, Hayes DN, Cancer Genome Atlas Research N. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell. 2010;17(1):98-110. PubMed PMID: 20129251; PMCID: PMC2818769. [CrossRef]
  280. Munoz L, Yeung YT, Grewal T. Oncogenic Ras modulates p38 MAPK-mediated inflammatory cytokine production in glioblastoma cells. Cancer Biol Ther. 2016;17(4):355-63. Epub 20160121. PubMed PMID: 26794430; PMCID: PMC4910925. [CrossRef]
  281. Aubin RG, Troisi EC, Montelongo J, Alghalith AN, Nasrallah MP, Santi M, Camara PG. Pro-inflammatory cytokines mediate the epithelial-to-mesenchymal-like transition of pediatric posterior fossa ependymoma. Nat Commun. 2022;13(1):3936. Epub 20220708. PubMed PMID: 35803925; PMCID: PMC9270322. [CrossRef]
  282. Delamarre E, Taboubi S, Mathieu S, Berenguer C, Rigot V, Lissitzky JC, Figarella-Branger D, Ouafik L, Luis J. Expression of integrin alpha6beta1 enhances tumorigenesis in glioma cells. Am J Pathol. 2009;175(2):844-55. Epub 20090702. PubMed PMID: 19574430; PMCID: PMC2716979. [CrossRef]
  283. Wu JS, Jiang J, Chen BJ, Wang K, Tang YL, Liang XH. Plasticity of cancer cell invasion: Patterns and mechanisms. Transl Oncol. 2021;14(1):100899. Epub 20201017. PubMed PMID: 33080522; PMCID: PMC7573380. [CrossRef]
  284. Joseph JV, Conroy S, Pavlov K, Sontakke P, Tomar T, Eggens-Meijer E, Balasubramaniyan V, Wagemakers M, den Dunnen WF, Kruyt FA. Hypoxia enhances migration and invasion in glioblastoma by promoting a mesenchymal shift mediated by the HIF1alpha-ZEB1 axis. Cancer Lett. 2015;359(1):107-16. Epub 20150112. PubMed PMID: 25592037. [CrossRef]
  285. Motevasseli M, Darvishi M, Khoshnevisan A, Zeinalizadeh M, Saffar H, Bayat S, Najafi A, Abbaspour MJ, Mamivand A, Olson SB, Tabrizi M. Distinct tumor-TAM interactions in IDH-stratified glioma microenvironments unveiled by single-cell and spatial transcriptomics. Acta Neuropathol Commun. 2024;12(1):133. Epub 20240816. PubMed PMID: 39148129; PMCID: PMC11328419. [CrossRef]
  286. Kraja FP, Jurisic VB, Hromic-Jahjefendic A, Rossopoulou N, Katsila T, Mirjacic Martinovic K, De Las Rivas J, Diaconu CC, Szoor A. Tumor-infiltrating lymphocytes in cancer immunotherapy: from chemotactic recruitment to translational modeling. Front Immunol. 2025;16:1601773. Epub 20250522. PubMed PMID: 40475782; PMCID: PMC12137109. [CrossRef]
  287. Wu J, Li R, Wang J, Zhu H, Ma Y, You C, Shu K. Reactive Astrocytes in Glioma: Emerging Opportunities and Challenges. Int J Mol Sci. 2025;26(7). Epub 20250323. PubMed PMID: 40243478; PMCID: PMC11989224. [CrossRef]
  288. Rodriguez SMB, Kamel A, Ciubotaru GV, Onose G, Sevastre AS, Sfredel V, Danoiu S, Dricu A, Tataranu LG. An Overview of EGFR Mechanisms and Their Implications in Targeted Therapies for Glioblastoma. Int J Mol Sci. 2023;24(13). Epub 20230705. PubMed PMID: 37446288; PMCID: PMC10341823. [CrossRef]
  289. An Z, Aksoy O, Zheng T, Fan QW, Weiss WA. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene. 2018;37(12):1561-75. Epub 20180111. PubMed PMID: 29321659; PMCID: PMC5860944. [CrossRef]
  290. Pournajaf S, Pourgholami MH. The mTOR pathway in Gliomas: From molecular insights to targeted therapies. Biomed Pharmacother. 2025;189:118237. Epub 20250621. PubMed PMID: 40544748. [CrossRef]
  291. Zhang Q, Zhao J, Xu T. Inhibition of eukaryotic initiation factor 4E by tomivosertib suppresses angiogenesis, growth, and survival of glioblastoma and enhances chemotherapy’s efficacy. Fundam Clin Pharmacol. 2023;37(4):807-15. Epub 20230207. PubMed PMID: 36691859. [CrossRef]
  292. Zhang C, Liang S, Zhang H, Wang R, Qiao H. Epigenetic regulation of mRNA mediates the phenotypic plasticity of cancer cells during metastasis and therapeutic resistance (Review). Oncol Rep. 2024;51(2). Epub 20231222. PubMed PMID: 38131215; PMCID: PMC10777459. [CrossRef]
  293. Etcheverry A, Aubry M, de Tayrac M, Vauleon E, Boniface R, Guenot F, Saikali S, Hamlat A, Riffaud L, Menei P, Quillien V, Mosser J. DNA methylation in glioblastoma: impact on gene expression and clinical outcome. BMC Genomics. 2010;11:701. Epub 20101214. PubMed PMID: 21156036; PMCID: PMC3018478. [CrossRef]
  294. Verma S, Goyal N, Goyal S, Kaur P, Gupta S. EZH2 Dysregulation and Its Oncogenic Role in Human Cancers. Cancers (Basel). 2025;17(19). Epub 20250924. PubMed PMID: 41097640; PMCID: PMC12523533. [CrossRef]
  295. Shi P, Hoang-Minh LB, Tian J, Cheng A, Basrai R, Kalaria N, Lebowitz JJ, Khoshbouei H, Deleyrolle LP, Sarkisian MR. HDAC6 Signaling at Primary Cilia Promotes Proliferation and Restricts Differentiation of Glioma Cells. Cancers (Basel). 2021;13(7). Epub 20210401. PubMed PMID: 33915983; PMCID: PMC8036575. [CrossRef]
  296. Zhang L, Yang Y, Li Y, Wang C, Bian C, Wang H, Wang F. Epigenetic regulation of histone modifications in glioblastoma: recent advances and therapeutic insights. Biomark Res. 2025;13(1):80. Epub 20250531. PubMed PMID: 40450300; PMCID: PMC12125905. [CrossRef]
  297. Yabo YA, Niclou SP, Golebiewska A. Cancer cell heterogeneity and plasticity: A paradigm shift in glioblastoma. Neuro-Oncology. 2022;24(5):669-82. PubMed PMID: WOS:000764685500001. [CrossRef]
  298. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, Dewhirst MW, Bigner DD, Rich JN. Glioma stem cells promote radioresistance by preferential activation of the DNA damage response. Nature. 2006;444(7120):756-60. Epub 20061018. PubMed PMID: 17051156. [CrossRef]
  299. Liu X, Chen Z, Yan P, Yang T, Zong D, Guo W, He X. Treatment mechanism and research progress of bevacizumab for glioblastoma. Am J Cancer Res. 2025;15(4):1874-901. Epub 20250425. PubMed PMID: 40371151; PMCID: PMC12070100. [CrossRef]
  300. Narsinh KH, Perez E, Haddad AF, Young JS, Savastano L, Villanueva-Meyer JE, Winkler E, de Groot J. Strategies to Improve Drug Delivery Across the Blood-Brain Barrier for Glioblastoma. Curr Neurol Neurosci Rep. 2024;24(5):123-39. Epub 20240405. PubMed PMID: 38578405; PMCID: PMC11016125. [CrossRef]
  301. Liu RN, Huang JH, Qi X, Pan Y, Wu E, Nizamutdinov D. Tumor Treating Fields and Combination Therapy in Management of Brain Oncology. Cancers (Basel). 2025;17(7). Epub 20250402. PubMed PMID: 40227773; PMCID: PMC11987984. [CrossRef]
  302. Yang T, Kong Z, Ma W. PD-1/PD-L1 immune checkpoint inhibitors in glioblastoma: clinical studies, challenges and potential. Hum Vaccin Immunother. 2021;17(2):546-53. Epub 20200709. PubMed PMID: 32643507; PMCID: PMC7899692. [CrossRef]
  303. Alomari O, Eyvazova H, Guney B, Al Juhmani R, Odabasi H, Al-Rawabdeh L, Mokresh ME, Erginoglu U, Keles A, Baskaya MK. Oncolytic Therapies for Glioblastoma: Advances, Challenges, and Future Perspectives. Cancers (Basel). 2025;17(15). Epub 20250801. PubMed PMID: 40805247; PMCID: PMC12346333. [CrossRef]
  304. Zhang C, Tan L, Zheng K, Xu Y, Wan J, Wu J, Wang C, Guo P, Feng Y. Single-cell/spatial integration reveals an MES2-like glioblastoma program orchestrated by immune communication and regulatory networks. Front Immunol. 2025;16:1699134. Epub 20251029. PubMed PMID: 41235227; PMCID: PMC12604987. [CrossRef]
  305. Weber GF. Molecular mechanisms of metastasis. Cancer Lett. 2008;270(2):181-90. Epub 20080605. PubMed PMID: 18522865. [CrossRef]
  306. Park JS, Kaygusuz Y, Kenum C, Gatie MI, He L, Sharma R, Chaligne R, Khan AG, Lorenz IC, Balderes PJ, Tammela T, Hadjantonakis AK, Ganesh K, Rudin CM, Massagué J. Emergence of metastasis-initiating cells during lung adenocarcinoma progression. Cancer Research. 2024;84(22). PubMed PMID: WOS:001373388900008. [CrossRef]
  307. Ocana OH, Corcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, Barrallo-Gimeno A, Cano A, Nieto MA. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer Cell. 2012;22(6):709-24. Epub 20121129. PubMed PMID: 23201163. [CrossRef]
  308. Huang M, Fu M, Wang J, Xia C, Zhang H, Xiong Y, He J, Liu J, Liu B, Pan S, Liu F. TGF-beta1-activated cancer-associated fibroblasts promote breast cancer invasion, metastasis and epithelial-mesenchymal transition by autophagy or overexpression of FAP-alpha. Biochem Pharmacol. 2021;188:114527. Epub 20210317. PubMed PMID: 33741330. [CrossRef]
  309. Ni Y, Zhou X, Yang J, Shi H, Li H, Zhao X, Ma X. The Role of Tumor-Stroma Interactions in Drug Resistance Within Tumor Microenvironment. Front Cell Dev Biol. 2021;9:637675. Epub 20210520. PubMed PMID: 34095111; PMCID: PMC8173135. [CrossRef]
  310. Zhang Y, Ding X, Zhang X, Li Y, Xu R, Li HJ, Zuo D, Chen G. Unveiling the contribution of tumor-associated macrophages in driving epithelial-mesenchymal transition: a review of mechanisms and therapeutic Strategies. Front Pharmacol. 2024;15:1404687. Epub 20240902. PubMed PMID: 39286635; PMCID: PMC11402718. [CrossRef]
  311. Li H, Qiu Z, Li F, Wang C. The relationship between MMP-2 and MMP-9 expression levels with breast cancer incidence and prognosis. Oncol Lett. 2017;14(5):5865-70. Epub 20170913. PubMed PMID: 29113219; PMCID: PMC5661385. [CrossRef]
  312. Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuna JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int J Mol Sci. 2020;21(24). Epub 20201220. PubMed PMID: 33419373; PMCID: PMC7767220. [CrossRef]
  313. Chen Q, Zou J, He Y, Pan Y, Yang G, Zhao H, Huang Y, Zhao Y, Wang A, Chen W, Lu Y. A narrative review of circulating tumor cells clusters: A key morphology of cancer cells in circulation promote hematogenous metastasis. Front Oncol. 2022;12:944487. Epub 20220818. PubMed PMID: 36059616; PMCID: PMC9434215. [CrossRef]
  314. Schaubaecher JB, Smiljanov B, Haring F, Steiger K, Wu Z, Ugurluoglu A, Luft J, Ballke S, Mahameed S, Schneewind V, Hildinger J, Canis M, Mittmann LA, Braun C, Zuchtriegel G, Kaiser R, Nicolai L, Mack M, Weichert W, Lauber K, Uhl B, Reichel CA. Procoagulant platelets promote immune evasion in triple-negative breast cancer. Blood. 2024;144(2):216-26. PubMed PMID: 38648571. [CrossRef]
  315. Nayak A, Warrier NM, Kumar P. Cancer Stem Cells and the Tumor Microenvironment: Targeting the Critical Crosstalk through Nanocarrier Systems. Stem Cell Rev Rep. 2022;18(7):2209-33. Epub 20220725. PubMed PMID: 35876959; PMCID: PMC9489588. [CrossRef]
  316. Parker AL, Cox TR. The Role of the ECM in Lung Cancer Dormancy and Outgrowth. Front Oncol. 2020;10:1766. Epub 20200911. PubMed PMID: 33014869; PMCID: PMC7516130. [CrossRef]
  317. Zhan Q, Liu B, Situ X, Luo Y, Fu T, Wang Y, Xie Z, Ren L, Zhu Y, He W, Ke Z. New insights into the correlations between circulating tumor cells and target organ metastasis. Signal Transduct Target Ther. 2023;8(1):465. Epub 20231221. PubMed PMID: 38129401; PMCID: PMC10739776. [CrossRef]
  318. Drolez A, Vandenhaute E, Delannoy CP, Dewald JH, Gosselet F, Cecchelli R, Julien S, Dehouck MP, Delannoy P, Mysiorek C. ST6GALNAC5 Expression Decreases the Interactions between Breast Cancer Cells and the Human Blood-Brain Barrier. Int J Mol Sci. 2016;17(8). Epub 20160811. PubMed PMID: 27529215; PMCID: PMC5000706. [CrossRef]
  319. Harris RE, Beebe-Donk J, Alshafie GA. Reduced risk of human lung cancer by selective cyclooxygenase 2 (COX-2) blockade: results of a case control study. Int J Biol Sci. 2007;3(5):328-34. Epub 20070613. PubMed PMID: 17589567; PMCID: PMC1893117. [CrossRef]
  320. Jekabsons MB, Merrell M, Skubiz AG, Thornton N, Milasta S, Green D, Chen T, Wang YH, Avula B, Khan IA, Zhou YD. Breast cancer cells that preferentially metastasize to lung or bone are more glycolytic, synthesize serine at greater rates, and consume less ATP and NADPH than parent MDA-MB-231 cells. Cancer Metab. 2023;11(1):4. Epub 20230220. PubMed PMID: 36805760; PMCID: PMC9940388. [CrossRef]
  321. Natarajan SK, Venneti S. Glutamine Metabolism in Brain Tumors. Cancers (Basel). 2019;11(11). Epub 20191024. PubMed PMID: 31652923; PMCID: PMC6893651. [CrossRef]
  322. Schwab A, Rao Z, Zhang J, Gollowitzer A, Siebenkas K, Bindel N, D’Avanzo E, van Roey R, Hajjaj Y, Ozel E, Armstark I, Bereuter L, Su F, Grander J, Bonyadi Rad E, Groenewoud A, Engel FB, Bell GW, Henry WS, Angeli JPF, Stemmler MP, Brabletz S, Koeberle A, Brabletz T. Zeb1 mediates EMT/plasticity-associated ferroptosis sensitivity in cancer cells by regulating lipogenic enzyme expression and phospholipid composition. Nat Cell Biol. 2024;26(9):1470-81. Epub 20240715. PubMed PMID: 39009641; PMCID: PMC11392809. [CrossRef]
  323. Xing Y, Ren ZQ, Jin R, Liu L, Pei JP, Yu K. Therapeutic efficacy and mechanism of CD73-TGFbeta dual-blockade in a mouse model of triple-negative breast cancer. Acta Pharmacol Sin. 2022;43(9):2410-8. Epub 20220126. PubMed PMID: 35082394; PMCID: PMC9433380. [CrossRef]
  324. Zhou Q, Meng Y, Li D, Yao L, Le J, Liu Y, Sun Y, Zeng F, Chen X, Deng G. Ferroptosis in cancer: From molecular mechanisms to therapeutic strategies. Signal Transduct Target Ther. 2024;9(1):55. Epub 20240308. PubMed PMID: 38453898; PMCID: PMC10920854. [CrossRef]
  325. Wang S, Johnson RM, Carozza JA, Fernandez D, Scicinski J, Verity NA, Mardjuki R, Cao X, Guo Y, Papkoff J, Ray N, Li L. ENPP1 inhibitor with ultralong drug-target residence time as an innate immune checkpoint blockade cancer therapy. Cell Rep Med. 2025;6(9):102336. Epub 20250905. PubMed PMID: 40914167; PMCID: PMC12490234. [CrossRef]
  326. Testa U, Pelosi E, Castelli G. Colorectal cancer: genetic abnormalities, tumor progression, tumor heterogeneity, clonal evolution and tumor-initiating cells. Med Sci (Basel). 2018;6(2). Epub 20180413. PubMed PMID: 29652830; PMCID: PMC6024750. [CrossRef]
  327. Massague J, Ganesh K. Metastasis-Initiating Cells and Ecosystems. Cancer Discov. 2021;11(4):971-94. PubMed PMID: 33811127; PMCID: PMC8030695. [CrossRef]
  328. Lovly CM, Iyengar P, Gainor JF. Managing Resistance to EFGR- and ALK-Targeted Therapies. Am Soc Clin Oncol Educ Book. 2017;37:607-18. PubMed PMID: 28561721; PMCID: PMC10183098. [CrossRef]
  329. Welch DR. Metastasis suppressors: a paradigm shift in cancer biology. Cancer Metastasis Rev. 2023;42(4):1057-9. PubMed PMID: 37535138; PMCID: PMC10772737. [CrossRef]
  330. Lens M, Schachter J. Immune Checkpoint Inhibitors in the Treatment of Advanced Melanoma in Older Patients: An Overview of Published Data. Cancers (Basel). 2025;17(11). Epub 20250530. PubMed PMID: 40507314; PMCID: PMC12153560. [CrossRef]
  331. Peng H, Li L, Zuo C, Chen MY, Zhang X, Myers NB, Hogg GD, DeNardo DG, Goedegebuure SP, Hawkins WG, Gillanders WE. Combination TIGIT/PD-1 blockade enhances the efficacy of neoantigen vaccines in a model of pancreatic cancer. Front Immunol. 2022;13:1039226. Epub 20221208. PubMed PMID: 36569934; PMCID: PMC9772034. [CrossRef]
  332. Hogg GD, Weinstein AG, Kingston NL, Liu X, Dres OM, Kang LI, Lander VE, Kao YL, Ahmad F, Knolhoff BL, Shenoy VV, Sells B, Jayasinghe RG, Houston A, Liu T, Herndon JM, Murphy KM, Ding L, Fields RC, Panni RZ, Hawkins WG, DeNardo DG. Combined Flt3L and CD40 agonism restores dendritic cell-driven T cell immunity in pancreatic cancer. Sci Immunol. 2025;10(110):eadp3978. Epub 20250815. PubMed PMID: 40815670. [CrossRef]
  333. I VC, Mustahidul Islam M, Bishnoi M, Priya S, Das Kurmi B, Koshey S, Patel P. Lifileucel Therapy for Metastatic Melanoma: Advancements in Tumor-infiltrating Lymphocyte-based Immunotherapy. Anticancer Agents Med Chem. 2025. Epub 20250704. PubMed PMID: 40621766. [CrossRef]
  334. Murad JP, Christian L, Rosa R, Ren Y, Buckley AJ, Lee EHJ, Lopez LS, Park AK, Yang J, Yamaguchi Y, Trac C, Adkins LN, Chang WC, Martinez C, June CH, Forman SJ, Ishihara J, Lee JK, Stern LA, Priceman SJ. Solid tumour CAR-T cells engineered with fusion proteins targeting PD-L1 for localized IL-12 delivery. Nat Biomed Eng. 2025. Epub 20251001. PubMed PMID: 41034514. [CrossRef]
  335. Huang T, Bei C, Hu Z, Li Y. CAR-macrophage: Breaking new ground in cellular immunotherapy. Front Cell Dev Biol. 2024;12:1464218. Epub 20241003. PubMed PMID: 39421021; PMCID: PMC11484238. [CrossRef]
  336. Kultti A, Li X, Jiang P, Thompson CB, Frost GI, Shepard HM. Therapeutic targeting of hyaluronan in the tumor stroma. Cancers (Basel). 2012;4(3):873-903. Epub 20120906. PubMed PMID: 24213471; PMCID: PMC3712709. [CrossRef]
  337. Zhao L, Pang Y, Zhou Y, Chen J, Fu H, Guo W, Xu W, Xue X, Su G, Sun L, Wu H, Zhang J, Wang Z, Lin Q, Chen X, Chen H. Antitumor efficacy and potential mechanism of FAP-targeted radioligand therapy combined with immune checkpoint blockade. Signal Transduct Target Ther. 2024;9(1):142. Epub 20240603. PubMed PMID: 38825657; PMCID: PMC11144707. [CrossRef]
  338. Cadieux B, Coleman R, Jafarinasabian P, Lipton A, Orlowski RZ, Saad F, Scagliotti GV, Shimizu K, Stopeck A. Experience with denosumab (XGEVA(R)) for prevention of skeletal-related events in the 10 years after approval. J Bone Oncol. 2022;33:100416. Epub 20220207. PubMed PMID: 35242510; PMCID: PMC8857591. [CrossRef]
  339. Zhou M, Liu C, Li B, Li J, Zhang P, Huang Y, Li L. Cell surface patching via CXCR4-targeted nanothreads for cancer metastasis inhibition. Nat Commun. 2024;15(1):2763. Epub 20240329. PubMed PMID: 38553476; PMCID: PMC10980815. [CrossRef]
  340. Mai Z, Lin Y, Lin P, Zhao X, Cui L. Modulating extracellular matrix stiffness: a strategic approach to boost cancer immunotherapy. Cell Death Dis. 2024;15(5):307. Epub 20240501. PubMed PMID: 38693104; PMCID: PMC11063215. [CrossRef]
  341. Qureshi Z, Altaf F, Khanzada M, Safi A, Asghar Z, Warraich D, Shah S. Liquid biopsies for early detection and monitoring of cancer: advances, challenges, and future directions. Ann Med Surg (Lond). 2025;87(6):3244-53. Epub 20250521. PubMed PMID: 40486554; PMCID: PMC12140778. [CrossRef]
  342. Abdullah KM, Sharma G, Singh AP, Siddiqui JA. Nanomedicine in Cancer Therapeutics: Current Perspectives from Bench to Bedside. Mol Cancer. 2025;24(1):169. Epub 20250609. PubMed PMID: 40490771; PMCID: PMC12147392. [CrossRef]
  343. Mohammadzadeh V, Atapour-Mashhad H, Shahvali S, Salehi B, Shaban M, Shirzad M, Salahvarzi A, Mohammadi M. Hydrogels as advanced drug delivery platforms for cancer immunotherapy: promising innovations and future outlook. J Nanobiotechnology. 2025;23(1):545. Epub 20250728. PubMed PMID: 40717098; PMCID: PMC12302882. [CrossRef]
  344. Stadtmauer EA, Fraietta JA, Davis MM, Cohen AD, Weber KL, Lancaster E, Mangan PA, Kulikovskaya I, Gupta M, Chen F, Tian L, Gonzalez VE, Xu J, Jung IY, Melenhorst JJ, Plesa G, Shea J, Matlawski T, Cervini A, Gaymon AL, Desjardins S, Lamontagne A, Salas-Mckee J, Fesnak A, Siegel DL, Levine BL, Jadlowsky JK, Young RM, Chew A, Hwang WT, Hexner EO, Carreno BM, Nobles CL, Bushman FD, Parker KR, Qi Y, Satpathy AT, Chang HY, Zhao Y, Lacey SF, June CH. CRISPR-engineered T cells in patients with refractory cancer. Science. 2020;367(6481). Epub 20200206. PubMed PMID: 32029687; PMCID: PMC11249135. [CrossRef]
  345. Shi Y, Zhang J, Li Y, Feng C, Shao C, Shi Y, Fang J. Engineered mesenchymal stem/stromal cells against cancer. Cell Death Dis. 2025;16(1):113. Epub 20250219. PubMed PMID: 39971901; PMCID: PMC11839947. [CrossRef]
  346. Zhang J, Jia Z, Pan H, Ma W, Liu Y, Tian X, Han Y, Wang Q, Zhou C, Zhang J. From induced pluripotent stem cell (iPSC) to universal immune cells: literature review of advances in a new generation of tumor therapies. Transl Cancer Res. 2025;14(4):2495-507. Epub 20250415. PubMed PMID: 40386273; PMCID: PMC12079212. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2026 MDPI (Basel, Switzerland) unless otherwise stated