Fact-checked by Grok 2 weeks ago

Tumor microenvironment

The tumor microenvironment (TME) is a dynamic and heterogeneous ecosystem surrounding malignant cells within a tumor, encompassing a diverse array of cellular and non-cellular elements that collectively regulate tumor initiation, growth, progression, and therapeutic resistance. This complex milieu includes tumor cells themselves, alongside non-malignant host components such as immune cells (e.g., T cells, macrophages, and myeloid-derived suppressor cells), stromal cells like cancer-associated fibroblasts (CAFs), endothelial cells forming vasculature, and the composed of proteins, glycoproteins, and proteoglycans. Additionally, soluble factors including cytokines, , growth factors, and metabolic byproducts permeate the TME, facilitating intercellular communication and shaping the tumor's local environment. The TME plays a pivotal role in cancer biology by providing structural support, nutrients, and oxygen through while also modulating immune responses to either promote anti-tumor immunity or foster that enables immune evasion. For instance, CAFs contribute to remodeling and secretion of pro-tumorigenic signals, enhancing tumor and , whereas tumor-associated macrophages often polarize toward an M2-like that supports and remodeling beneficial to the cancer. within the TME, driven by rapid tumor outpacing vascular supply, further activates pathways like hypoxia-inducible factor (HIF)-1α, which upregulates genes for , , and . Beyond its influence on development, the TME is crucial for metastatic dissemination, as pre-metastatic niches in distant organs are conditioned by tumor-derived exosomes and factors that recruit bone marrow-derived cells to prepare sites for colonization. Therapeutically, the TME poses significant barriers to treatments like and ; for example, dense can limit drug penetration, while immunosuppressive cells such as regulatory T cells (Tregs) and PD-L1-expressing components dampen T-cell activation. Recent advances highlight the TME's , with targeting strategies—such as normalizing vasculature or reprogramming immune cells—emerging as promising avenues to improve outcomes in solid tumors.

Fundamentals

Definition and Components

The tumor microenvironment (TME) is defined as a complex, dynamic surrounding malignant tumor cells, encompassing both cellular and non-cellular elements derived from the host that interact bidirectionally to influence tumor , progression, , , and therapeutic resistance. Unlike the tumor , which consists primarily of neoplastic cells, the TME actively modulates cancer behavior through reciprocal signaling, evolving from an initially passive supportive role to an active driver of oncogenesis by fostering conditions that promote tumor survival and immune evasion. This microenvironment is stimulated by tumor-secreted factors, leading to molecular, cellular, and physical alterations in the surrounding , including , , and stiffness, which collectively sustain the neoplastic niche. The core components of the TME can be categorized into cellular and non-cellular elements. Cellular components include non-malignant cells such as endothelial cells forming the vascular network, fibroblasts (including cancer-associated fibroblasts), adipocytes, and various immune cells like macrophages, T lymphocytes, B cells, natural killer cells, neutrophils, and dendritic cells, all of which can exhibit pro-tumorigenic or anti-tumorigenic functions depending on their and activation state. Non-cellular components comprise the (), composed of structural proteins like , , , and that can constitute up to 60% of the tumor mass and regulate , migration, and signaling, as well as soluble factors such as cytokines, growth factors (e.g., VEGF, TGF-β), , and extracellular vesicles (EVs) including exosomes that mediate intercellular communication. Emerging elements, such as metabolic gradients (e.g., depletion and waste accumulation) and microbial influences, further contribute to TME complexity by shaping tumor adaptation. The TME exhibits significant heterogeneity across cancer types, with distinct compositions in solid tumors—characterized by dense stromal and vascular elements supporting localized invasion—compared to hematologic malignancies, where the niche predominates with mesenchymal stromal cells providing soluble factors like to nurture leukemic cells. This variability arises from tumor-specific genetic and epigenetic alterations, anatomical location, and evolutionary pressures, resulting in immune landscapes ranging from inflamed (T-cell infiltrated) to excluded or desert-like (immune-silent). Over time, the TME transitions from a passive scaffold to an active participant in oncogenesis, reprogramming host cells to sustain tumor growth and suppress anti-tumor immunity. For instance, the TME can recapitulate embryonic developmental niches by reactivating pathways like Wnt, , and , which promote stemness and self-renewal in cancer cells, mimicking the supportive stroma of embryogenesis to create a protective haven for tumor propagation.

Historical Development

The concept of the tumor microenvironment (TME) originated in 19th-century observations linking host tissues to cancer development. In 1863, hypothesized that cancer arises from loci of chronic inflammation, based on his pathological examinations revealing leukocytes infiltrating neoplastic tissues and suggesting that irritants at these sites trigger neoplastic transformation. This insight first connected non-malignant host responses to tumorigenesis. Building on this, Stephen Paget's 1889 "seed and soil" hypothesis explained patterns of observed in 735 breast cancer autopsies, positing that tumor cells (seeds) preferentially colonize compatible organ microenvironments (soil) rather than disseminating randomly.70201-8/fulltext) Interest in the TME revived in the mid-20th century with mechanistic studies on tumor-host interactions. Judah Folkman's 1971 theory of tumor proposed that beyond a minimal size, solid tumors require inducing from host to sustain growth, identifying vascular dependence as a rate-limiting step controlled by the TME. In the , experiments using recombination in models revived focus on stromal influences; Gerald Cunha and colleagues demonstrated that mammary could instruct epithelial and either promote or inhibit neoplastic progression depending on its composition, underscoring bidirectional epithelial-stromal signaling in cancer. From the to , the TME emerged as a core paradigm in cancer biology. Mina Bissell and co-workers in 2002 articulated the TME as an active organizer of tumor architecture, integrating and stromal cues that dictate malignant behavior beyond genetic alterations in cancer cells alone. This view was formalized in Douglas Hanahan and Robert Weinberg's "" framework (2000, revised 2011), which added TME-enabled traits like sustaining proliferative signaling through and as hallmarks of malignancy.81683-9)00127-9) Entering the 2010s, integrative models incorporated immune evasion and metabolic reprogramming within the TME, fueled by advances revealing suppressive immune dynamics. By the 2020s, single-cell sequencing has illuminated TME plasticity, mapping heterogeneous cellular states and interactions at unprecedented resolution to inform therapeutic targeting.

Vascular Microenvironment

Endothelial Cells and Angiogenesis

Endothelial cells form the inner lining of blood vessels within the tumor microenvironment, providing structural support and facilitating nutrient and oxygen delivery to sustain tumor growth. These cells are highly responsive to angiogenic signals, particularly (VEGF), which binds to VEGF receptors on their surface to initiate intracellular signaling cascades that promote endothelial , survival, and . This signaling drives key processes such as , where specialized tip cells lead the extension of new vascular branches, and tube formation, enabling the assembly of functional networks. Tumor angiogenesis is a hypoxia-driven process that enables the formation of new blood vessels from pre-existing ones to meet the metabolic demands of expanding tumors. Under low oxygen conditions, tumor cells release pro-angiogenic factors like VEGF and fibroblast growth factor (FGF), which activate endothelial cells to initiate the angiogenic cascade. The process unfolds in sequential steps: first, degradation of the basement membrane by matrix metalloproteinases allows endothelial cells to invade the surrounding extracellular matrix; this is followed by endothelial migration and proliferation guided by chemotactic gradients; finally, the nascent vessels undergo maturation, involving recruitment of pericytes and stabilization of the structure. Despite these mechanisms, tumor vasculature exhibits profound abnormalities that impair efficient and exacerbate the hostile microenvironment. Newly formed vessels are often tortuous and dilated, with irregular branching patterns that lead to heterogeneous blood flow and regions of inadequate oxygenation. A hallmark feature is their leakiness, resulting from disrupted endothelial junctions and fenestrations, which causes plasma and elevated interstitial pressure. Additionally, these vessels suffer from deficient pericyte coverage, as fail to adequately ensheath endothelial tubes, contributing to structural instability and further permeability issues. The concept of tumor angiogenesis was pioneered by , who in 1971 isolated a tumor angiogenesis factor (TAF) from solid tumors, demonstrating its mitogenic effect on endothelial cells and establishing angiogenesis as a critical rate-limiting step in tumor progression. This discovery laid the foundation for targeting angiogenesis therapeutically, exemplified by , a that neutralizes VEGF and inhibits endothelial activation, thereby suppressing new vessel formation in various cancers. 's approval in 2004 marked the first clinically validated anti-angiogenic therapy, improving in tumors reliant on VEGF-driven vascularization, though resistance mechanisms often emerge over time.

Hypoxia

Hypoxia in the tumor microenvironment (TME) arises primarily from the rapid proliferation of tumor cells that outpaces the development of vascular supply, leading to regions of oxygen deprivation. This imbalance creates two main types of hypoxic zones: , where oxygen gradients form due to limited from nearby vessels to distant tumor cells, and perfusion-limited hypoxia, characterized by total or intermittent oxygen deprivation resulting from inadequate flow through abnormal tumor vasculature. These conditions are prevalent in many tumors, with approximately 50-60% exhibiting hypoxic regions due to this supply-demand mismatch. Under hypoxic conditions, hypoxia-inducible factors (HIFs), particularly HIF-1α, are stabilized by inhibiting the activity of prolyl hydroxylase domain enzymes that normally target it for degradation in normoxia. Stabilized HIF-1α dimerizes with HIF-1β and translocates to the nucleus, where it binds hypoxia response elements to transcribe target genes involved in adaptation. Key targets include those promoting glycolysis, such as glucose transporter 1 (GLUT1) and lactate dehydrogenase A (LDHA), which facilitate anaerobic metabolism; vascular endothelial growth factor (VEGF), which drives angiogenesis; and matrix metalloproteinases (MMPs), which enhance tumor invasion. This transcriptional program enables tumor cells to survive and proliferate in low-oxygen environments. The consequences of extend to profound tumor adaptations, including a metabolic shift to the Warburg effect, where cells preferentially perform aerobic even in the presence of oxygen, sustaining energy production and biomass under nutrient stress. also exerts selective pressure, favoring the survival and dominance of aggressive, apoptosis-resistant clones that can better tolerate oxygen deprivation and promote . Furthermore, hypoxic regions contribute to radio- and resistance by limiting (ROS) generation, which are essential for the oxidative damage induced by these treatments. briefly triggers angiogenic responses via VEGF to improve vascularization, though this often results in dysfunctional vessels that exacerbate the hypoxic milieu. Recent studies in the 2020s have linked HIF pathway dysregulation in hypoxic niches to immunotherapy failure, as stabilized HIF-1α upregulates immunosuppressive factors like PD-L1, impairing T-cell infiltration and effector function in tumors such as melanoma and lung cancer.

Enhanced Permeability and Retention Effect

The enhanced permeability and retention (EPR) effect refers to the passive accumulation of macromolecules and nanoparticles in solid tumors due to abnormal vascular structure and function. First described in 1986 by Matsumura and Maeda, the phenomenon was observed when radiolabeled proteins selectively accumulated in tumor tissues compared to normal organs, attributing this to tumor-specific pathophysiological features.90004-1) This effect has since become a foundational concept in oncology for explaining how nanotherapeutics can achieve higher intratumoral concentrations without active targeting. The mechanism of the EPR effect involves two key components: enhanced permeability of tumor vasculature and prolonged retention within the tumor . Tumor-induced , driven by (VEGF), disrupts endothelial cell junctions, creating leaky gaps ranging from 100 to 2000 nm in diameter—far larger than the <5 nm gaps in normal vessels. These gaps allow macromolecules and nanoparticles (typically >5 kDa or >10 nm) to extravasate from the bloodstream into the tumor extracellular space. Retention occurs due to impaired or absent lymphatic drainage in tumors, which prevents efficient clearance of these agents, leading to their prolonged presence and gradual release. Several factors influence the magnitude of the EPR effect across tumors. It is more pronounced in highly vascularized tumors, such as sarcomas, where dense, immature vessel networks facilitate greater , compared to hypovascular or desmoplastic tumors like pancreatic cancers, which exhibit reduced permeability due to dense stromal barriers. design plays a critical role, with optimal sizes of 20-100 nm promoting efficient through the leaky while minimizing rapid renal clearance; larger particles (>200 nm) may face steric hindrance, and smaller ones (<10 nm) are often cleared too quickly. In drug delivery, the EPR effect underpins several approved nanomedicines by enabling selective tumor accumulation and reduced systemic toxicity. A prime example is pegylated liposomal doxorubicin (Doxil), approved by the FDA in 1995 for Kaposi's sarcoma, which exploits EPR-mediated extravasation to deliver the chemotherapeutic payload directly to tumor sites, improving efficacy and tolerability over free doxorubicin. Despite these successes, clinical translation has revealed limitations, including heterogeneous EPR across patients and tumor regions, which can result in suboptimal drug delivery and diminished nanotherapy efficacy. Recent analyses in 2024 have highlighted this variability, emphasizing that factors like tumor stage, interstitial pressure, and patient comorbidities often lead to only modest (1-2 fold) enhancements in accumulation compared to preclinical models.

Stromal Microenvironment

Carcinoma-Associated Fibroblasts

Carcinoma-associated fibroblasts (CAFs) represent a dominant stromal cell population within the (TME), characterized by their activation from quiescent fibroblasts and their multifaceted contributions to tumor progression. These cells arise primarily from local tissue-resident fibroblasts, such as pancreatic stellate cells, but can also originate from pericytes that differentiate and recruit to the tumor site or from bone marrow-derived mesenchymal progenitors that undergo transdifferentiation. In response to signals from cancer cells, CAFs become activated through pathways involving (TGF-β) and (PDGF), which induce expression of α-smooth muscle actin (α-SMA) and promote a contractile phenotype. This activation process, often termed stromal activation, enables CAFs to sustain a supportive niche for tumor growth and metastasis across various carcinomas, including . Single-cell RNA sequencing (scRNA-seq) analyses conducted in the 2020s have revealed significant heterogeneity among CAFs, delineating distinct subtypes with varying functional roles. Myofibroblastic CAFs, marked by high α-SMA expression, predominate in fibrotic tumors and drive matrix stiffening through contractility. In contrast, inflammatory CAFs secrete interleukin-6 (IL-6) and chemokines like CXCL12, fostering an inflammatory milieu that supports tumor expansion. These subtypes exhibit pro-tumorigenic properties in most contexts, such as enhancing invasion in breast and pancreatic cancers, though subsets like antigen-presenting CAFs may exert anti-tumor effects by modulating immunity in specific settings. This plasticity underscores the context-dependent nature of CAF phenotypes, as evidenced by scRNA-seq profiles from over 8,000 cells in pancreatic ductal adenocarcinoma. CAFs exert tumor-promoting functions through the secretion of growth factors, including hepatocyte growth factor () and fibroblast growth factors (), which stimulate cancer cell proliferation and angiogenesis via paracrine signaling. They also produce extracellular matrix () components like collagen and periostin, contributing to desmoplasia and tumor stiffness, while their actin-myosin contractility generates mechanical forces that facilitate cancer cell invasion. Recent studies highlight metabolic coupling between CAFs and cancer cells, where CAFs recycle lactate shuttled from glycolytic tumor cells to fuel their own fibrogenic and immunosuppressive activities, as demonstrated in 2023 research on pancreatic cancer models. This lactate-dependent symbiosis enhances deposition by CAFs, reinforcing the desmoplastic barrier in the .

Extracellular Matrix Remodeling

The extracellular matrix (ECM) in the tumor microenvironment (TME) is a dynamic network primarily composed of fibrillar collagens such as types I and III, which provide structural integrity and tensile strength to the tissue. Fibronectin and laminins contribute to cell adhesion and signaling, while proteoglycans like versican regulate matrix hydration and growth factor sequestration. Hyaluronic acid (HA), a glycosaminoglycan, further enhances hydration and creates a hydrated scaffold that supports tumor cell proliferation and motility. Remodeling of the ECM is driven by upregulated matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, which degrade collagen and other components to facilitate tumor invasion. Lysyl oxidase (LOX) and its family members promote collagen crosslinking, increasing matrix density and stiffness. In pancreatic ductal adenocarcinoma, this remodeling manifests as desmoplasia, a dense fibrotic stroma that compresses blood vessels and hinders drug delivery. Carcinoma-associated fibroblasts contribute to this process by secreting ECM proteins, amplifying the remodeling effects. Biomechanically, ECM stiffening activates integrin-mediated signaling through focal adhesion kinase (FAK), promoting epithelial-to-mesenchymal transition (EMT) and enhanced tumor cell invasion. In three-dimensional (3D) matrix models, increased matrix stiffness induces force-dependent cell migration, where tumor cells exert traction forces that correlate with metastatic potential. These changes create a permissive environment for tumor progression by altering mechanotransduction pathways. Strategies to target the ECM include hyaluronidases, such as pegvorhyaluronidase alfa (PEGPH20), which degrade excess HA in high-HA tumors like pancreatic ductal adenocarcinoma to improve drug penetration. These have been investigated in clinical trials, including a phase III study in hyaluronan-high metastatic pancreatic cancer that increased objective response rates but did not improve overall survival as of 2020. Recent preclinical research as of 2024 explores hyaluronidase inhibitors, such as delphinidin, to suppress cancer metastasis by preserving ECM integrity. Additionally, 2025 studies highlight potential in inducing a "stromal switch" to reprogram pro-tumorigenic stromal components for enhanced therapeutic efficacy.

Immune Microenvironment

Innate Immune Cells

Innate immune cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and neutrophils, are rapidly recruited to the tumor microenvironment (TME) and often adopt pro-tumor phenotypes that support cancer progression. TAMs, derived from circulating monocytes, are the most abundant innate immune population in many solid tumors and typically polarize toward an M2-like state under the influence of cytokines such as IL-4 and IL-13, which activate the IL-4Rα/STAT6 pathway to promote immunosuppressive and tissue-remodeling functions. MDSCs, identified in mice as Gr-1+CD11b+ immature myeloid cells, expand in response to tumor-derived factors like GM-CSF and exhibit heterogeneous subsets that inhibit anti-tumor immunity. Neutrophils, the first responders to inflammatory cues, can polarize to an N2 pro-tumor phenotype driven by TGF-β signaling, shifting from potential anti-tumor activity to one that fosters tumor growth and dissemination.00215-3) Recruitment of these cells into the TME is orchestrated by chemokines and growth factors secreted by tumor and stromal cells. Monocytes are primarily attracted by (also known as ), which binds on their surface to facilitate infiltration and differentiation into , enhancing tumor colonization in models of breast and prostate cancer. Once in the TME, promote angiogenesis by secreting (), which stimulates endothelial cell proliferation and new vessel formation, thereby supplying nutrients and oxygen to the expanding tumor mass. Neutrophils contribute to extracellular matrix () remodeling through the release of (), particularly , which degrades basement membranes and facilitates tumor invasion by creating pathways for cancer cell migration. The pro-tumor shift in these cells involves metabolic and enzymatic mechanisms that suppress anti-tumor responses while aiding tumor progression. M2-polarized TAMs express high levels of arginase-1 (ARG1), which depletes L-arginine in the local milieu, thereby inhibiting nitric oxide production and cytotoxic activity in neighboring immune effectors. Similarly, MDSCs upregulate ARG1 and inducible nitric oxide synthase (iNOS), leading to L-arginine depletion that impairs T cell receptor signaling and proliferation through cyclin D3 downregulation and cell cycle arrest. Neutrophils in the N2 state release neutrophil extracellular traps (NETs), web-like structures of DNA and proteins that capture and promote tumor cell adhesion to distant sites; recent 2025 studies in mouse models of neuroendocrine prostate cancer liver metastasis demonstrate that serotonin-induced NETs enhance metastatic seeding by remodeling the hepatic ECM and evading immune clearance. These functions collectively create an immunosuppressive niche that sustains tumor growth and metastasis.

Adaptive Immune Cells

Tumor-infiltrating lymphocytes (TILs) represent a critical component of the adaptive immune response within the tumor microenvironment (TME), encompassing primarily , , B cells, and regulatory T cells (Tregs). serve as the main effectors, directly targeting and lysing tumor cells, while orchestrate broader immune activation by supporting dendritic cell maturation and enhancing through cytokine signaling. B cells contribute by producing tumor-specific antibodies and presenting antigens to T cells, fostering localized immune hubs such as tertiary lymphoid structures. In contrast, Tregs often exert immunosuppressive effects, though their roles can vary by context. The extent of TIL infiltration varies significantly across tumors, classifying them as "hot" or "cold" based on immune cell density. Hot tumors exhibit high TIL infiltration, particularly of CD8+ T cells, correlating with enhanced antitumor immunity and better prognosis in cancers like melanoma and breast carcinoma. Cold tumors, conversely, show sparse TIL presence, often due to physical and biochemical barriers such as dense extracellular matrix (ECM) components like collagen and hyaluronan, which impede lymphocyte migration into the tumor core. Abnormal vascular adhesion molecules on endothelial cells further restrict T cell extravasation, perpetuating an immunosuppressive niche. In antitumor activity, CD8+ T cells recognize tumor-specific neoantigens presented on major histocompatibility complex class I (MHC-I) molecules via their T cell receptors, triggering perforin- and granzyme-mediated cytotoxicity. This recognition activates cytokine release, notably interferon-gamma (IFN-γ), which amplifies MHC-I expression on tumor cells and recruits additional immune effectors to the TME. CD4+ T cells bolster this process by secreting IL-2 and promoting Th1 polarization, while B cells enhance efficacy through antibody-dependent cellular cytotoxicity. However, prolonged antigen exposure leads to T cell exhaustion, marked by upregulation of inhibitory receptors such as PD-1 and TIM-3, which dampen proliferative and cytotoxic functions. Recent advances in spatial transcriptomics have illuminated TIL organization in responsive tumors; for instance, a 2024 study using spatial proteomics and single-cell transcriptomics in metastatic melanoma revealed that clinical responses to TIL adoptive therapy correlate with preexisting CD8+ T cell-myeloid networks, indicating clustered TIL distributions predictive of therapeutic success. B cells exhibit dual functionality, with some subsets promoting tumor progression via IL-10 secretion, while others drive antibody-mediated immunity. Tregs contribute to suppression by depleting IL-2 and releasing TGF-β, countering effector responses in the TME.

Immunosuppressive Mechanisms

The tumor microenvironment (TME) fosters immunosuppression through multiple interconnected mechanisms that enable cancer cells to evade immune surveillance. These include the upregulation of immune checkpoints, metabolic alterations, and cytokine-mediated signaling that collectively inhibit effector T cell function and promote regulatory immune cell activity. Such processes are driven by tumor cells, stromal components, and recruited immune cells, creating a barrier to effective antitumor immunity. Molecular pathways play a central role in TME-induced immunosuppression. The PD-1/PD-L1 axis is frequently upregulated on tumor cells and infiltrating immune cells, where PD-L1 expression on tumor cells binds PD-1 on activated T cells, delivering inhibitory signals that attenuate T cell proliferation and cytokine production.30432-3) Similarly, the enzyme indoleamine 2,3-dioxygenase (IDO) is expressed by tumor and antigen-presenting cells in the TME, catalyzing the catabolism of tryptophan into kynurenine, which depletes local tryptophan levels and starves T cells of this essential amino acid, thereby impairing their activation and survival. Cellular crosstalk further amplifies these effects, with immunosuppressive cells like regulatory T cells (Tregs), tumor-associated macrophages (TAMs), and myeloid-derived suppressor cells (MDSCs) contributing key mediators. Tregs, often enriched in the TME, secrete transforming growth factor-β (TGF-β), which suppresses effector T cell differentiation and promotes apoptosis in cytotoxic lymphocytes. TAMs, polarized toward an M2-like phenotype, release interleukin-10 (IL-10), an anti-inflammatory cytokine that inhibits dendritic cell maturation and dampens Th1 responses essential for antitumor immunity. MDSCs, in turn, generate reactive oxygen species (ROS) through NADPH oxidase activity, creating oxidative stress that disrupts T cell receptor signaling and induces T cell apoptosis. These interactions, involving both innate and adaptive immune components, form a suppressive network that sustains tumor progression. Microenvironmental factors exacerbate this immunosuppression by altering the biochemical milieu. The Warburg effect in tumor cells leads to excessive glycolysis and lactate production, acidifying the TME to a pH below 6.8, which impairs T cell motility, cytokine secretion, and glycolytic metabolism required for effector functions.00213-7) Additionally, the ectoenzyme on tumor and immune cells hydrolyzes extracellular AMP to adenosine, which accumulates in the hypoxic TME and signals through A2A and A2B receptors on T cells to suppress their proliferation and interferon-γ production.30388-9) Recent 2025 studies have demonstrated that combined checkpoint blockade, such as dual PD-1 and CTLA-4 inhibition, can partially overcome TME-induced T cell exhaustion in non-small cell lung cancer (NSCLC) by reinvigorating exhausted CD8+ T cells and reducing suppressive cytokine levels in the TME.

Additional Components

Metabolic Factors

The tumor microenvironment (TME) is characterized by metabolic reprogramming, prominently featuring the , where cancer cells preferentially undergo aerobic glycolysis, leading to excessive lactate production and extracellular acidosis with a pH typically ranging from 6.5 to 6.8. This lactate accumulation not only supports tumor bioenergetics but also creates a hostile milieu that impairs immune cell function. A key dynamic is the competition for glucose between tumor cells and infiltrating immune cells, such as T lymphocytes, where tumors deplete available glucose, restricting effector T cell proliferation, glycolytic capacity, and cytokine production like IFN-γ. Metabolic symbiosis within the TME further exacerbates these imbalances, as carcinoma-associated fibroblasts (CAFs) undergo glycolytic reprogramming to produce lactate, which is shuttled to oxidative tumor cells via monocarboxylate transporters (uptake in tumors) and (export from CAFs), fueling tumor growth while acidifying the surroundings. Complementing this, many tumors exhibit glutamine addiction, relying on glutamine as a critical nitrogen and carbon source for biosynthesis, proliferation, and redox balance, often outcompeting stromal and immune cells in the nutrient-scarce TME. Hypoxia serves as a metabolic trigger in this context, inducing adaptive responses that amplify these dependencies. These metabolic alterations have profound impacts on TME homeostasis, including the upregulation of hypoxia-inducible genes like PKM2, a pyruvate kinase isoform that promotes glycolytic flux and HIF-1α stabilization independent of direct HIF mediation, thereby sustaining tumor anabolic demands. Additionally, in tumors with mutant isocitrate dehydrogenase (IDH), the oncometabolite 2-hydroxyglutarate (2-HG) accumulates, inhibiting α-ketoglutarate-dependent dioxygenases and driving T cell anergy, myeloid-derived suppressor cell expansion, and overall immunosuppression in the TME. Recent advances in 2024 have enhanced metabolic imaging of TME heterogeneity through PET tracers beyond FDG, such as those targeting glutamine uptake (e.g., 18F-(2S,4R)-4-fluoroglutamine) and lactate dynamics, enabling non-invasive visualization of nutrient gradients and therapeutic responses. Therapeutic targeting of these pathways has been explored in clinical trials, such as the phase 2 CANTATA trial of glutaminase inhibitor telaglenastat (CB-839) combined with cabozantinib in advanced renal cell carcinoma, which did not demonstrate significant improvements in progression-free survival. As of 2025, further studies in combinations like nivolumab have shown good tolerability but limited efficacy across cohorts.

Extracellular Vesicles

Extracellular vesicles (EVs) serve as key mediators of intercellular communication within the tumor microenvironment (TME), facilitating the exchange of bioactive molecules between tumor cells and surrounding stromal and immune components. These nanoscale membrane-bound structures, secreted by various cell types including cancer cells, carry diverse cargo such as proteins, lipids, and nucleic acids that modulate TME dynamics, promoting tumor progression and immune evasion. Tumor-derived EVs are particularly enriched in oncogenic factors, enabling them to reprogram recipient cells and establish supportive niches. EVs in the TME are primarily classified into two major types based on their biogenesis and size: exosomes and microvesicles. Exosomes, ranging from 30 to 150 nm in diameter, originate from the endosomal pathway through inward budding of multivesicular endosomes (MVEs), while microvesicles, typically 100 to 1000 nm, form by outward budding and fission from the plasma membrane. Tumor-derived EVs, including both subtypes, are often enriched with microRNAs (miRNAs) and proteins that reflect the altered metabolic and signaling states of cancer cells, such as oncogenic miRNAs like miR-21 and matrix metalloproteinases. The biogenesis and release of EVs involve intricate cellular machinery. For exosomes, the endosomal sorting complex required for transport (ESCRT) pathway facilitates cargo sorting into MVEs, with subsequent fusion to the plasma membrane mediated by Rab GTPases, such as Rab27a and Rab27b, enabling release into the extracellular space. Microvesicles, in contrast, rely on cytoskeletal elements like actin and ARF6 for budding. Once released, EVs are taken up by recipient cells through mechanisms including endocytosis, phagocytosis, or direct membrane fusion, allowing cargo transfer that alters cellular behavior in the TME. Tumor-derived EVs exert profound functional effects by enabling horizontal transfer of genetic material and proteins. For instance, exosomes carrying miR-21 from cancer cells or associated macrophages can be internalized by endothelial cells, upregulating vascular endothelial growth factor (VEGF) expression and thereby promoting angiogenesis to support tumor vascularization. Additionally, these EVs precondition distant metastatic sites by recruiting bone marrow-derived cells and remodeling the extracellular matrix, creating a fertile "pre-metastatic niche" that facilitates tumor dissemination. Recent advancements highlight the diagnostic and therapeutic potential of EVs in the TME. As of 2025, research has advanced EV-based liquid biopsies for non-invasive profiling of TME components, leveraging their cargo to detect tumor heterogeneity and immune status with high specificity in patient plasma samples. In preclinical models, engineered EVs loaded with small interfering RNAs () targeting oncogenic pathways, such as , have demonstrated efficacy in suppressing tumor growth and enhancing immunotherapy responses by modulating TME interactions. These developments underscore EVs' dual role as both pathological drivers and promising therapeutic vectors.

Roles in Cancer Progression

Tumor Growth and Invasion

The tumor microenvironment (TME) plays a pivotal role in promoting primary tumor expansion through paracrine signaling loops involving cancer-associated fibroblasts (CAFs). CAFs secrete hepatocyte growth factor (HGF), which activates the c-Met receptor on tumor cells, thereby enhancing proliferation, migration, and epithelial-mesenchymal transition (EMT) via downstream ERK1/2 and STAT3 pathways in gastric and hepatocellular carcinomas. This HGF/c-Met axis fosters tumor-initiating cell plasticity and chemoresistance, amplifying growth signals in a non-cell-autonomous manner. Mechanical signaling from the extracellular matrix (ECM) further drives tumor growth by altering cellular mechanics. Increased ECM stiffness, often reaching 4–10 kPa in tumors compared to <1 kPa in normal tissues, activates mechanotransducers such as , , and , promoting proliferation through and cytoskeletal remodeling in hepatocellular and lung cancers. Stiffness-induced YAP nuclear translocation upregulates genes like and , sustaining a pro-growth phenotype. Invasion at the primary site is facilitated by collective cell migration, where tumor cells move as cohesive clusters guided by TME components like tumor-associated macrophages (TAMs). TAMs induce a CD44high state in leader tumor cells via CCL8 secretion, driving cohesive detachment and multicellular strand invasion in breast carcinomas, as observed in organotypic cultures and models. Podocalyxin (PODXL), a sialomucin upregulated in invasive tumors, enhances protrusion formation by binding and promoting dynamics at the , thereby increasing motility and invasiveness in pancreatic ductal cells. Tumor cells exhibit plasticity in motility modes, switching between mesenchymal and amoeboid types to navigate the TME. Mesenchymal motility involves elongated cells with integrin-dependent adhesions and -mediated degradation, proceeding at 0.1–0.5 μm/min, while amoeboid motility features rounded, bleb-driven movement via Rho/ contractility at speeds up to 25 μm/min, allowing squeezing through pores without . Transitions between these modes, such as mesenchymal-to-amoeboid (), enable adaptive invasion in response to density. Feedback loops between tumor cells and the TME perpetuate growth and invasion through recruitment of supportive elements. Tumor-derived factors like sonic hedgehog (SHh) and TGF-β activate pancreatic stellate cells, inducing desmoplastic reactions with excessive deposition that stiffens the stroma and promotes further recruitment in . Similar loops occur in , where neoplastic cells stimulate granulin-expressing stromal cells to enhance accumulation and , fostering a pro-invasive niche. Intravital imaging studies from 2023 have revealed real-time co-evolution of the TME and tumor during invasion, demonstrating how TAMs co-migrate with EGFR+ cancer cells in breast models, dynamically remodeling the ECM to support collective advance.

Metastasis and Therapy Resistance

The tumor microenvironment (TME) plays a pivotal role in preparing distant sites for metastasis by forming the pre-metastatic niche, where extracellular vesicles (EVs) and exosomes from primary tumors "educate" secondary organs such as the lungs. Tumor-derived exosomes, containing proteins, mRNAs, and microRNAs like miR-122, reprogram glucose metabolism in target tissues and recruit macrophages to foster an inflammatory environment conducive to tumor cell colonization. For instance, in lung metastasis models, exosomes enriched in S100A8/A9 heterodimers activate NF-κB signaling in myeloid-derived suppressor cells (MDSCs), promoting their recruitment and immune suppression at the pre-metastatic site. This S100A8/A9-mediated process, induced by cytokines like TNF-α and TGF-β from endothelial cells, creates a permissive niche that enhances vascular permeability and myeloid cell infiltration, facilitating subsequent tumor dissemination. Epithelial-mesenchymal transition () in the TME further drives metastatic potential through signals that induce cellular plasticity. TGF-β secreted by cancer-associated fibroblasts and immune cells in the TME activates transcription factors such as and , which repress E-cadherin expression and promote mesenchymal traits like invasiveness and migration. This TGF-β-driven pathway operates via both transcriptional upregulation of Snail/Slug and post-transcriptional mechanisms, including miRNA downregulation, enabling tumor cells to detach from the primary site. Hybrid epithelial/mesenchymal (E/M) states, stabilized by EMT transcription factors like and Zeb1, confer enhanced plasticity, allowing cells to adapt to circulatory stress and seed distant metastases while retaining partial epithelial features for efficient colonization. These intermediate states are enriched in circulating tumor cells and correlate with poor prognosis in cancers like and colorectal. The TME also contributes to therapy resistance by creating protective niches that shield tumor cells from chemotherapeutic agents. Hypoxic cores within solid tumors, arising from inadequate vascularization, limit drug penetration due to fibrosis and abnormal perfusion while upregulating hypoxia-inducible factor-1α (HIF-1α), which induces ATP-binding cassette (ABC) transporters like (PGP) and multidrug resistance protein 1 (MRP1) to efflux drugs and reduce intracellular accumulation. This hypoxic adaptation further promotes and inhibits pathways, such as those involving BNIP3, allowing cancer cells to survive treatment-induced stress. Tumor-associated macrophages (TAMs), particularly M2-polarized ones, exacerbate resistance by secreting chemokines like CCL17 and CCL22, which activate the CCR4 receptor on tumor cells, leading to endoplasmic reticulum stress and GRP78-mediated translocation of MRP1 to the membrane for enhanced drug efflux. In , this TAM-tumor cell interaction via the CCL17/CCL22–CCR4–ATF6–GRP78 axis directly confers resistance to 5-fluorouracil by increasing its extrusion. Recent multi-omics analyses in 2025 have linked specific TME signatures to elevated metastasis risk in , highlighting immune exhaustion and adhesion dysregulation. Proteogenomic profiling of metastatic versus non-metastatic tumors revealed a 16-gene network in the TME, including downregulated (e.g., , CCL19) associated with reduced T and infiltration, alongside upregulated selectins (SELE, SELL) tied to epithelial-mesenchymal transition, with phosphorylated RPS6 (p-RPS6) emerging as a key prognostic marker for poor outcomes. Additionally, adaptive following anti-PD-1 involves TME-driven T exhaustion, characterized by upregulated inhibitory checkpoints like LAG-3 and on CD8+ T cells, coupled with remodeling that impedes immune infiltration. These mechanisms underscore the TME's role in post-immunotherapy evasion, as seen in non-small where resistant tumors exhibit decreased TCF-1 expression and altered myeloid dynamics.

Research and Applications

Modeling Approaches

In vitro models provide foundational platforms for studying tumor microenvironment (TME) interactions by simulating cellular components in controlled settings. Two-dimensional (2D) co-cultures, such as those involving tumor cells and cancer-associated fibroblasts, enable the examination of direct cell-cell communications and soluble factor exchanges that drive tumor progression, though they often fail to replicate . These models are cost-effective and high-throughput, facilitating initial screens of TME modulators like signaling. Three-dimensional (3D) organoids and spheroids advance beyond 2D systems by incorporating () components and hypoxic gradients, which mimic the avascular tumor and promote realistic , , and metabolic adaptations observed . Tumor spheroids, formed via hanging drop or low-adherence techniques, recapitulate nutrient diffusion limitations and oxygen gradients, leading to central and enhanced akin to solid tumors. Patient-derived organoids further personalize these models by preserving genetic and stromal features from biopsies, allowing longitudinal studies of TME evolution. Advanced models integrate dynamic physiological cues to better emulate TME complexity. Tumor-on-a-chip platforms utilize to simulate vascular flow, , and interstitial gradients, enabling real-time observation of endothelial-tumor interactions and immune . Innovations in 2024 have incorporated perfusable vascular networks within 3D matrices, facilitating studies of and under biomimetic conditions. Patient-derived xenografts (PDXs), where human tumor fragments are implanted into immunodeficient mice, partially recapitulate the human TME with stromal elements preserved in early passages (1-2) before replacement by murine stroma; immune elements are absent in standard PDXs but can be incorporated via humanized models using engrafted human hematopoietic stem cells, offering a bridge between and fidelity for personalized therapy testing. In vivo models provide systemic context for TME research, capturing host-tumor crosstalk in living organisms. Transgenic mouse models, such as the MMTV-PyMT strain for , express polyoma middle T antigen under the mouse mammary tumor virus promoter, recapitulating multistage tumorigenesis with progressive TME remodeling including activation and immune infiltration. These models exhibit high of mammary tumors and lung , enabling genetic manipulations to dissect TME roles in progression. embryos serve as optically transparent hosts for xenografting human tumor cells, allowing high-resolution intravital of dynamics, vascular invasion, and niche interactions without ethical constraints of mammalian models. By 2025, the integration of (AI) with spatial technologies has enabled predictive modeling of TME heterogeneity, where algorithms analyze multiplexed imaging and transcriptomics to forecast immune responses and therapeutic outcomes from patient samples. Despite these advances, mouse models exhibit key discrepancies with TME, including divergent immune cell compositions and stromal responses that limit translational accuracy.

Genetic Influences

Host germline variants, particularly single nucleotide polymorphisms (SNPs) in immune-related genes, significantly influence the composition and functionality of the tumor microenvironment (TME). For instance, specific HLA class I alleles have been associated with enhanced tumor-infiltrating lymphocyte (TIL) infiltration, promoting a more immunogenic TME that correlates with improved patient outcomes in various cancers. Similarly, germline variants in cytokine loci, such as those in the IL6 gene, can modulate inflammatory responses within the TME; the IL6 rs1800795 polymorphism, for example, has been linked to altered IL-6 production and increased cancer susceptibility through exacerbated chronic inflammation. These genetic factors highlight how inherited variations in immune regulation shape the recruitment and activation of immune cells in the TME. Beyond immune genes, germline variants in vascular and () components directly impact TME architecture. SNPs in the VEGFR2 (KDR) gene, such as rs2071559, have been shown to affect receptor signaling, leading to dysregulated that enhances tumor vascularization and nutrient supply while also promoting hypoxic niches conducive to immune evasion. In the , variants in COL1A1, a major gene, predispose to fibrotic remodeling; for example, certain COL1A1 polymorphisms contribute to increased collagen deposition, creating dense stromal barriers that hinder immune cell penetration and delivery in fibrotic tumor niches. These effects underscore the role of host genetics in sculpting physical and vascular elements of the TME that influence tumor growth and therapeutic access. In cancer-specific contexts, germline mutations in and genes alter stromal responses within the TME, often leading to heightened immune activation due to genomic instability. BRCA1/2 carriers exhibit increased stromal infiltration of pro-inflammatory cells and elevated expression of immune checkpoints, which can paradoxically enhance responses in and ovarian cancers by creating a more inflamed TME. Recent genome-wide association studies (GWAS) from the 2020s have further linked variants to TME traits and ; for instance, a 2023 GWAS identified SNPs in immune modifier genes that predict TIL abundance and survival across multiple tumor types, emphasizing their prognostic value. Additionally, 2024 investigations into PDCD1 polymorphisms, such as rs2227981, have revealed their modulation of PD-1 expression on T cells, influencing TME and thereby affecting efficacy in and patients.

Therapeutic Strategies

Therapeutic strategies targeting the tumor microenvironment (TME) aim to disrupt immunosuppressive and supportive elements that foster cancer progression, enhancing the efficacy of conventional treatments. Vascular targeting represents a cornerstone of these approaches, with anti-angiogenic agents like inhibiting (VEGF) to prune abnormal tumor vasculature and limit nutrient supply. Approved by the FDA in 2004 for advanced , bevacizumab has since been integrated into regimens for various solid tumors, where it modulates the TME by reducing and immune evasion. Complementing this, vascular normalization agents such as cediranib, a , restore more functional blood vessels, improving tumor and oxygenation in glioblastoma patients. Clinical studies have shown that cediranib-induced increases in correlate with enhanced when combined with chemoradiation, highlighting its role in alleviating TME barriers to . Stromal and immune components of the TME are addressed through inhibitors of cancer-associated fibroblasts () and . TGF-β blockers like galunisertib (LY2157299), a small-molecule of TGF-β receptor I, target CAF activation and remodeling that shield tumors from immune attack. In preclinical models, galunisertib disrupts TGF-β-driven CAF-tumor crosstalk, potentiating immune responses in pancreatic and breast cancers. Immune checkpoint inhibitors, such as , a PD-1 approved by the FDA in 2014 for unresectable or metastatic , reinvigorate T-cell activity within the immunosuppressive TME. By blocking PD-1 interactions, pembrolizumab alters the TME to favor antitumor immunity across multiple indications, including non-small cell . Emerging stromal-immune strategies include chimeric antigen receptor () T-cell therapies engineered against tumor-associated macrophages (), which remain preclinical as of 2025. These CAR-T cells, targeting TAM markers like CD206, reprogram the myeloid compartment to reduce and enhance T-cell infiltration in solid tumors. Emerging modalities expand TME targeting to metabolic reprogramming, vesicular transport, and viral disruption. Metabolic drugs such as IDH1/2 inhibitors (e.g., ) block mutant enzymes that produce oncometabolite 2-hydroxyglutarate, thereby normalizing TME metabolism and sensitizing immune cells in IDH-mutant gliomas. These agents alter the immunosuppressive metabolic landscape, improving outcomes in low-grade gliomas when used perioperatively. Extracellular vesicle ()-based delivery systems leverage tumor-derived EVs for targeted drug transport, exploiting their natural to penetrate the TME and deliver chemotherapeutics directly to cancer cells. Oncolytic viruses like (T-VEC), an engineered approved by the FDA in 2015 for advanced , selectively lyse tumor cells while releasing tumor antigens to remodel the TME and stimulate systemic immunity. T-VEC disrupts immunosuppressive elements, including and regulatory T cells, enhancing efficacy. As of 2025, bispecific antibodies targeting the TME-tumor interface, such as ivonescimab engaging PD-1 and VEGF to simultaneously block immune suppression and , are in advanced clinical development with planned FDA biologics license application submissions in late 2025 for tumors. therapies integrating these agents with checkpoint inhibitors or chemotherapeutics have demonstrated superior in overcoming TME-mediated , as seen in trials where dual blockade reduces adaptive immune evasion in non-small cell . Such multimodal approaches, including CAR-T with metabolic inhibitors, address heterogeneous TME dynamics to improve durable responses.