Next Article in Journal
Facile Synthesis of Polysubstituted Pyridines via Metal-Free [3+3] Annulation Between Enamines and β,β-Dichloromethyl Peroxides
Previous Article in Journal
What Is Similar, What Is Different? Characterization of Mitoferrin-like Proteins from Arabidopsis thaliana and Cucumis sativus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The 3D Language of Cancer: Communication via Extracellular Vesicles from Tumor Spheroids and Organoids

by
Simona Campora
* and
Alessandra Lo Cicero
*
Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, 90128 Palermo, Italy
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(15), 7104; https://doi.org/10.3390/ijms26157104
Submission received: 23 June 2025 / Revised: 16 July 2025 / Accepted: 22 July 2025 / Published: 23 July 2025
(This article belongs to the Special Issue Recent Advances in 3D Tumor Models for Cancer Research)

Abstract

Extracellular vesicles (EVs) have emerged as key mediators of intercellular communication, gaining recognition as tumor biomarkers and promising therapeutic targets. As the study of EVs advances, it has become increasingly clear that the cellular context in which they are produced significantly influences their composition and function. Traditional two-dimensional in vitro models are being progressively replaced by more advanced three-dimensional systems, such as tumor spheroids and organoids. These 3D models are particularly valuable in cancer research, providing a more accurate representation of the complex cellular and molecular heterogeneity that characterizes tumors, better mimicking the in vivo microenvironment compared to standard monolayer cultures. This review explores the role of EVs derived from tumor spheroids and organoids in key oncogenic processes, including tumor growth, metastasis, and interactions within the tumor microenvironment. We highlight how EVs contribute to the spread of cancer cells, affecting surrounding tissues, and promote immune evasion, which poses significant challenges in cancer therapy.

1. Introduction

Extracellular vesicles (EVs) are ubiquitous, membrane-bound nanosized particles involved in various physiological and pathological processes [1,2]. They play crucial roles in intercellular communication by delivering bioactive cargos, such as proteins, lipids, and nucleic acids, that can significantly influence target recipient cells, organs, and the surrounding microenvironment [3,4]. Due to these properties, EVs are being explored as promising biomarkers, therapeutic agents, and Drug Delivery Systems (DDSs).
Although EVs play essential homeostatic functions, they are also implicated in different diseases, including cancer, which is a major cause of worldwide mortality. According to the World Health Organization, nearly 20 million new cancer diagnoses and 10 million cancer-related deaths were reported in 2023, with cases continuing to increase annually. Many studies have reported that cancer cells secrete higher quantities of EVs compared to non-malignant cells. These vesicles are actively involved in promoting tumor progression by facilitating invasion, metastasis, angiogenesis, and immune evasion [5,6].
Although monolayer cell cultures are widely employed for isolating EVs due to their affordability and workability, they fail to accurately replicate the structural and functional complexity, resulting in EVs that lack authentic pathological characteristics observed in vivo [7,8]. Tumor masses consist of cancer cells and immune and stromal cells, producing a typical dense extracellular matrix (ECM), which influences EV production and composition [9]. Therefore, a more representative model for isolating EVs was investigated by developing different 3D cell culture models, which often also include ECM components. For this purpose, both adherent and non-adherent approaches have been explored for growing cells in 3D structures [10]. Adherent methods included the use of scaffolds (natural or synthetic polymers, hydrogels, decellularized tissues), microfluidics, and bioreactors [11]. While these models offer advantages, including long-term culture, homogeneous aggregates, and controlled cell number and viability, they also present some disadvantages, including difficulties in isolating cells and EVs and removing residual serum, resulting in the contamination of serum-derived vesicles, which complicates the purification of cell-specific EVs. These limitations can be overcome by non-adherent techniques, such as hanging drop and liquid overlay techniques, which form tumor spheroids and organoids. Spheroids effectively mimic key tumor characteristics, including cell–cell and cell–matrix interactions, the nutrients and oxygen gradients, and the heterogeneity typical of solid tumors. On the other hand, organoids are more complex systems that include additional vascular components and are enriched with more representative extracellular compounds. The choice of the appropriate 3D system depends on the specific aim and application.
This review provides a comprehensive analysis of the molecular composition and functional roles of EVs released by tumor spheroids and organoids. It highlights their involvement in tumor growth, their role in modulating the tumor microenvironment, and their potential as biomarkers and therapeutic targets in cancer treatment.

2. Biogenesis and Characteristics of Extracellular Vesicles

EVs are mainly generated through two mechanisms: the blebbing of the plasma membrane (microvesicles) or within multivesicular bodies (MVBs), followed by release by exocytosis as exosomes [12,13]. Multiple mechanisms contribute to vesicle formation, influencing their heterogeneity, size, and function. Exosomes and microvesicles consist of a mixture of membrane and cytosolic elements, including proteins, lipids, and RNAs, and their composition varies based on their biogenesis site [1,14] (Figure 1).
Exosomes are the smallest type of EVs, typically ranging from 30 to 150 nanometers in diameter. They originate from the endosomal system through the inward budding of the plasma membrane, leading to the formation of early endosomes, which mature into late endosomes. These can follow two different pathways: they may fuse with lysosomes, resulting in the degradation of their contents, or they may give rise to MVBs [15]. During MVB maturation, the endosomal membrane undergoes inward budding, forming mature intraluminal vesicles (ILVs). Different molecular cargoes are selectively sorted into ILVs in the late endosomes through specific signaling pathways and molecular machinery. In this context, the Endosomal Sorting Complex Required for Transport (ESCRT) plays a crucial role in ILV biogenesis [16,17], comprising several protein subcomplexes (ESCRT-0, ESCRT-I, ESCRT-II, and ESCRT-III) that sequentially assemble on the endosomal membrane to facilitate cargo trafficking and vesicle budding [16,17,18]. Additionally, sorting molecules such as tetraspanins and specific lipids like ceramides assist in cargo selection [19], while transmembrane proteins, including CD9, CD63, and CD81, contribute to the organization of microdomains on the endosomal membrane [20,21,22]. MVBs ultimately fuse with the plasma membrane to release the ILVs as exosomes into the extracellular space. This process is regulated by several factors, including Rab GTPases (e.g., Rab27a and Rab27b), soluble NSF (N-ethylmaleimide-Sensitive Factor) attachment receptor (SNARE) proteins, and other regulatory factors [23,24].
Microvesicles (MVs) are typically larger than exosomes, ranging from 100 to 1000 nanometers in diameter. They are formed through the direct outward budding and fission of the plasma membrane, a process that involves the rearrangement of lipid and protein components [13,14,15]. This remodeling is often driven by phospholipid redistribution, such as the externalization of phosphatidylserine, which plays a pivotal role in membrane curvature and budding during both exosome and microvesicle formation [19,25]. Additionally, the reorganization of the actin cytoskeleton, regulated by small GTPases like ARF6 and RhoA, is essential in this process. Specifically, these small molecules modulate the contractile machinery necessary for MV fission and release into the extracellular environment [26].
Both exosomes and MVs are complex structures carrying a wide variety of biomolecules, reflecting the status and function of their originating cells. The membranes of EVs have a specific lipidic composition that is rich in phospholipids like phosphatidylserine, phosphatidylcholine, and phosphatidylethanolamine, and is involved in membrane curvature and budding. Moreover, sphingomyelin and ceramides contribute to membrane structure and dynamics [27,28], while cholesterol is involved in maintaining the stability and fluidity of the EV membrane [29]. Furthermore, the EV membrane contains tetraspanins (e.g., CD9, CD63, CD81), integrins, and various receptors crucial for their formation, release, targeting, and uptake by recipient cells.
On the other hand, the inner molecular composition typically includes proteins, lipids, nucleic acids, heat shock proteins (HSP70, HSP90), cytoskeletal proteins (actin, tubulin), and enzymes (GAPDH, enolase), contributing to the vesicle function and intercellular communication abilities. For instance, EVs can carry proteins involved in signaling pathways, including kinases, transcription factors, and growth factors, which modulate cellular responses in recipient cells [20,21,22,30]. Additionally, EVs can transport different classes of RNA, including messenger RNAs (mRNA) translated into proteins in recipient cells, potentially influencing their gene expression profile [31,32]; microRNAs, like small and non-coding RNAs, which regulate gene expression post-transcriptionally and are involved in processes such as cell proliferation, differentiation, and apoptosis [33,34,35,36,37]; and long non-coding RNAs (lncRNAs) and small nucleolar RNAs (snoRNAs), which may also modulate gene expression and cellular functions [38,39]. EVs can also contain fragments of double-stranded DNA (dsDNA) reflecting the genetic material of the parent cell [40,41]. Furthermore, mitochondrial DNA (mDNA) can be packaged and transferred into exosomes, potentially impacting the metabolic and signaling pathways of recipient cells [42,43]. Moreover, mitochondria can also release exosome-like vesicles themselves [44].
Although the process of EV biogenesis is similar in healthy and tumor cells [45], the molecular composition can significantly depend on the donor cell type. For instance, tumor-derived EVs can transfer oncogenic factors like mutated proteins or miRNA that contribute to tumor growth [46,47], invasion [48], formation of pre-metastatic niches [49], and metastasis [50]. Furthermore, these EVs can also carry molecules associated with drug resistance [51,52] and immuno evasion [53], promoting a lack of therapeutic efficacy and tumor progression.

3. Isolation and Characterization Techniques

To study and exploit the potential role of different EVs, several steps, including isolation, purification, and characterization, are required [54,55]. Accurate and reliable techniques are crucial for separating distinct EV populations and ensuring the purity and integrity of the vesicles for downstream applications, such as proteomic, genomic, and functional analyses [56]. Various methods have been employed for this purpose, each with its advantages and limitations [57,58,59]. Differential ultracentrifugation, often regarded as the gold standard for the isolation of EVs [60,61], involves sequential centrifugation steps at increasing speeds to remove cells, debris, and large vesicles, followed by ultracentrifugation at high speeds to pellet EVs. Density gradient purification, which utilizes a continuous density gradient of sucrose or iodixanol in combination with differential centrifugation, achieves higher purity levels for isolating EVs, which may be combined with other methods, such as nanomembrane-based filtration [62]. Since EVs contain protein makers indicative of their cellular origin, specific EV subtypes can be collected by targeting peculiar surface proteins using immunoaffinity-based approaches [62,63]. For instance, common biomarkers for EV isolation include tetraspanins like CD9, CD63, and CD81.
After isolation and enrichment, EVs can be characterized in terms of size and concentration using techniques like Nanoparticle Tracking Analysis (NTA) [64] and Transmission Electron Microscopy (TEM) [65,66]. Their molecular cargo can be further investigated through methods like flow cytometry [67,68], mass spectrometry [69,70], and RNA-seq datasets for miRNAs, lncRNAs, and mRNAs [71].

4. Tumor-Derived EVs

EVs play a pivotal role in intercellular communication under both physiological and pathological conditions [72]. Cancer cells produce and release higher quantities of EVs compared to normal cells, often containing altered cargo specifically adapted to support tumor heterogeneity and adaptability. The impact of EVs is highly context-dependent, varying with tumor type, stage, and microenvironmental conditions. In many cancers, EVs facilitate proliferation, invasion, angiogenesis, and metastasis. These vesicles disseminate molecular signals both locally and systemically, contributing to the maintenance and progression of the primary tumor mass [73,74]. Numerous studies have demonstrated EVs’ ability to carry bioactive molecules, promoting cancer development and progression [37,75,76]. Within the tumor microenvironment, EVs released by various cells —such as cancer cells, immune cells, and stromal cells—contribute to oncogenic processes, including tumor growth, angiogenesis, immune evasion, and metastasis [33,77,78,79,80,81,82] (Figure 2).
EVs contribute to cancer progression by promoting the formation of pre-metastatic niches (PMNs), a specialized microenvironment that prepares for the implantation of organ-specific metastasis [83]. EVs can alter the vasculature and accumulation of bone marrow-derived cells (BMDCs) to enhance metastatic colonization, as observed in melanoma [84]. Tumor-derived EVs have been shown to establish immunosuppressive PMNs by impairing Natural Killer (NK) cell activity and inhibiting the maturation of dendritic cells (DC). García-Silva and colleagues demonstrated that melanoma-secreted EVs promote lymphangiogenesis and lymph node metastasis through the nerve growth factor (NGF) receptor (NGFR), which acts on lymphatic endothelial cells [85]. A tumor mass is a complex of different types of cells, including cancer cells, carcinoma-associated fibroblasts (CAFs), adipocytes, stem cells, endothelial cells, and immune cells [86]. EVs modulate tumor vasculature by delivering pro-angiogenic factors, such as vascular endothelial growth factor (VEGF) [87], and upregulating the expression of the vascular endothelial growth factor receptor FLT1 [81] in endothelial cells, which stimulates angiogenesis, enhances vascular permeability, and facilitates the formation of pre-metastatic niches. These vascular changes enable key steps in the metastatic process, including tumor cell intravasation, extravasation, and metastatic colonization. Moreover, CAF-derived EVs in particular play a critical role in PMN formation, as suggested by Kong et al. They demonstrated that CAF EV uptake by lung fibroblasts is mediated by integrin α2β1, a fundamental process for cell activation, PMN development, and metastasis formation [87]. However, multiple other integrin subtypes are also implicated in EV-driven tumor progression. EVs exhibit tissue-specific accumulation patterns that correlate with future metastatic sites; notably, EVs expressing α6β4 and α6β1 integrins are linked to lung metastasis, while those bearing αvβ5 integrin preferentially target the liver [81]. Moreover, EVs released by cancer stem cells differ in molecular composition from those released from differentiated cancer cells. This has been exemplified in the triple-negative breast cancer (TNBC) model in which EVs secreted by cancer stem cells drive PMN remodeling in both in vitro and in vivo systems [88].
Additionally, EVs can modulate the immune system by suppressing immune cell activity, thereby creating an immunosuppressive tumor microenvironment [89,90]. EVs also contribute to remodeling the extracellular matrix by altering the surrounding stromal cells’ phenotypes, making them more supportive of cancer cell proliferation and invasion. For instance, cancer cell-derived EVs can reprogram fibroblasts into CAFs, further promoting tumor progression [91,92,93]. A notable example of this mechanism is provided by Yang and colleagues, who demonstrated the crucial role of colorectal cancer (CRC)-derived EVs in the early formation of liver metastasis. Their study revealed that transforming growth factor-beta 1 (TGF-β1) carried by CRC-derived EVs promotes the differentiation of hepatic stellate cells (HSCs) into CAFs. These CAFs recruit myeloid-derived suppressor cells (MDSCs), which inhibit NK activity and establish immunosuppressive PMNs [94].
Furthermore, EVs are implicated in drug resistance, making cells more resilient to conventional therapies [95]. In response to classic chemotherapy, cancer cells, like melanoma cells, secrete more EVs able to restart cancer growth in in vivo systems, specifically by enhancing arginase 1 and IL10 in stromal cells and stimulating the transcription of genes involved in the DNA repair process [96]. EVs also contribute to the drug efflux mechanism, as reported by Yang et al., who demonstrated that EVs promote the expulsion of the temozolomide (TMZ) in glioblastoma (GBM) cells, thereby reducing its cytotoxic efficacy and inducing drug resistance [97]. Programmed cell death protein-ligand 1 (PD-L1) found on the surface of tumor EVs inhibits T cell activation, leading to immune escape mechanisms mediating tumor progression [90,98,99].
Even EVs released by other cancer-associated cells, including CAF or immune cells, play a pivotal role in this process [100,101]. In thyroid cancer, programmed cell death protein 1 (PD-1) therapy resistance is mediated by tumor-associated macrophage (TAM)- derived EVs through the delivery of miR-21-5p, which inhibits methyltransferase-like 3 (METTL3) [102].
On the other hand, EVs represent potential targets for innovative therapeutic strategies due to their crucial role in the intricate network of interactions that drive cancer’s development and progression. Furthermore, they can be functionalized as a drug delivery system by incorporating drugs and antibody-binding moieties specific to the fragment crystallizable (Fc) domain for active tumor targeting [103,104]. For instance, biomarkers, which are measurable indicators of physiological and pathological biological processes, are involved in tracking disease progression and response to treatment. EVs from cancer cells hold significant promise for diagnostics, as they carry specific molecules—such as mutated DNA, proteins, and microRNAs—that serve as biomarkers, reflecting the state and function of their originating cells and, therefore, indicating the presence and progression of cancer, including metastasis [32,105,106]. For instance, the proteins PF4 and AACT in serum EV samples and the two transmembrane proteins CD147 and A33 in fecal EVs are key markers for colorectal cancer [107,108]. By offering an efficient tool for tumor detection, EVs present several advantages, including cost-effectiveness, pain reduction, and the potential to serve as a non-invasive alternative to traditional surgical methods.

5. Tumor Spheroids and Organoids: An In Vitro Model

Tumor spheroids and organoids are three-dimensional (3D) cell culture models that provide more physiologically relevant insights into tumor biology than traditional two-dimensional (2D) monolayer cultures [109]. Spheroids are typically derived from single-cell suspensions with self-assembly capabilities to mimic the architecture and microenvironment of solid tumors. This includes replicating complex cellular interactions, cell–matrix interactions, and nutrient and oxygen gradients present in vivo [110,111]. On the other hand, organoids are more complex 3D structures that closely mimic native tumor architecture and function, typically comprising multiple cell types, including elements of the tumor microenvironment (TME) [112].
Spheroids exhibit an outer proliferative cell layer, an intermediate quiescent zone, and a hypoxic core caused by restricted nutrient and oxygen diffusion (Figure 3) [113,114,115]. This layered formation creates microenvironments within the spheroid, resulting in oxygen, nutrients, and metabolite gradients affecting cellular behavior and drug response. In particular, the hypoxic core provides a platform to explore how reduced oxygen levels influence tumor progression and resistance to therapy [116,117]. These properties make spheroids an invaluable tool for investigating cancer features, progression, drug responses, and therapeutic resistance. The same architecture is also evident in some organoids, depending on the type (generally cancerous ones), size (big organoids), and culture conditions [118].
Spheroids and organoids can be synthesized using liquid-based techniques that prevent cell adhesion to the substrate while promoting cell–cell interactions. The size of the spheroids depends on the initial number of cells. Several methods can be used to culture cells in 3D. Static cultures in ultra-low-attachment plates or flasks are coated with materials such as poly (2-hydroxyethyl methacrylate) (pHEMA), hyaluronic acid (HA), poly-d-lysine, laminin, or agarose to prevent substrate adhesion [119,120]. The Hanging Drop method, on the other hand, exploits gravity to promote cell aggregation within a drop of culture medium suspended from a surface. In dynamic suspension cultures, the constant movement of the cell suspension within the culture medium prevents substrate adhesion and favors cell–cell contacts. In this context, spinner flasks—devices that maintain the suspension through mechanical stirring—and the RCCS/RWV system, a rotating wall vessel bioreactor, are commonly used [121]. In magnetic levitation, cells are incubated with magnetic iron oxide nanoparticles (MIO NPs), which are taken up through endocytosis. The subsequent application of a magnetic field induces cellular aggregation in plates, leading to spheroid formation [122]. Alternatively, spheroids can also be generated directly in the presence of porous microspheres that act as scaffolds and are simply added to the culture medium [123].
Unlike monolayer cells, which grow on flat surfaces and fail to replicate the complex interactions and cellular diversity found in tumors, spheroids and organoids maintain proper cell–cell and cell–matrix interactions. Even the proteomic profile varies when comparing 2D and 3D systems of the same cells. For instance, spheroids of primary breast cancer showed an increase in collagen and matrix metalloproteinase (MMP) expression compared with monolayer cultures, suggesting a more articulated and organized extracellular environment [124]. On the other hand, epigenetic patterns, including methylation and microRNA expression, change significantly between the two systems [125]. This distinction makes them more suitable for accurate tumor behavior modeling.
Furthermore, solid tumors are a complex ecosystem composed not only of cancer cells but also of non-cancerous cells that significantly contribute to tumor microenvironment (TME) formation and tumor progression. These include immune cells, cancer-associated fibroblasts (CAFs), endothelial cells (ECs), pericytes, adipocytes, and neurons (Figure 3). Although the TME cell composition depends on the tumor stage, the type of cancer, and the patient origin, it is well established that these cells play a role in tumor pathogenesis and their crosstalk with cancer cells through secreted molecules carried by vesicles [126]. In this scenario, multicellular tumor spheroids (MTSs), also called assembloids, provide a more realistic model by incorporating different cell types into a 3D structure [127]. These co-culture systems enable a more comprehensive understanding of tumor–immune interactions and the impact of the tumor microenvironment on cancer progression. For instance, pancreatic ductal adenocarcinoma (PDAC) organoids have been used as a model for studying the cytokine-mediated signaling pathways involved in CAF cell activity [128]. Similarly, MTSs of the osteosarcoma cell line and mesenchymal stromal cells (MSCs) have provided insights into cytokine-driven ECM deposition in osteosarcoma [129]. Moreover, more complex spheroids of pancreatic cancer cells, fibroblasts, and endothelial cells are an optimal system to investigate mechanisms of in vivo-like drug resistance [130].
Organoids are 3D miniature structures derived from various stem or tissue sources: embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), adult tissue-resident stem cells (ASCs), and cancer-derived cells [131]. Based on their cellular origin, organoids can be broadly classified into two categories: tissue-derived organoids, which originate from direct isolation of stem cells from primary tissues, and organoids generated from somatic or pluripotent stem cells through directed differentiation protocols.
Organoids derived from ESCs or iPSCs are particularly valuable for modeling early human development, including organogenesis and congenital disorders. Those derived from adult stem cells retain many characteristics of their tissue of origin, making them suitable for studying organ-specific physiology, regeneration, and disease pathology [132].
Cancer-derived organoids, especially those established from patient samples, enable researchers to model tumor heterogeneity, genetic mutations, and drug responses in a personalized context. A major focus in current organoid research lies in patient-derived organoids (PDOs), particularly those established from tumor biopsies [133]. PDOs closely preserve the histological architecture, molecular features, and functional behaviors of the original tumors, providing a powerful system for studying tumor biology, genetic heterogeneity, and therapeutic response in a patient-specific context.
Despite their promise, the inherent self-organizing nature of organoids presents challenges in achieving uniformity and reproducibility. As a result, ongoing research is focused on refining culture conditions to produce consistent and stable organoid models for clinical and translational applications [134].
Most tumor organoid cultures currently use Matrigel as an ECM scaffold [135]. However, synthetic matrix alternatives, such as synthetic hydrogels and gelatin methacrylate (GelMA), are increasingly being explored, offering greater control over chemical composition and mechanical properties to enhance the reproducibility and scalability of organoid culture [136,137]. One of the main challenges in developing tumor organoids is to selectively promote tumor cell proliferation while inhibiting the growth of non-tumor cells present in heterogeneous cell suspensions. To address this, careful optimization of the culture medium is essential. This typically involves adjusting the composition of cytokines and growth factors to create a selective environment that favors the survival and expansion of tumor cells [138,139].
A major limitation of 3D spheroid cultures is the development of diffusion gradients as spheroid size increases. Once spheroids exceed 500 μm in diameter, their multilayered architecture restricts the transport of oxygen and nutrients while impairing the clearance of metabolic waste [140]. This frequently leads to central necrosis and compromises overall structural integrity and cellular viability. Despite these well-known physiological constraints, critical experimental parameters—such as oxygen levels, fetal bovine serum (FBS) concentration, media composition (e.g., glucose and calcium content), and initial cell density—are often inconsistently reported or neglected, contributing to poor reproducibility and hindering clinical translation. For instance, spheroids cultured at low oxygen levels (3%) showed reduced size and increased necrotic areas, while FBS concentrations above 10% promoted more compact spheroids with distinct proliferative and necrotic zones [141]. These findings underscore the sensitivity of 3D models to environmental conditions.
Although in vitro culture systems inherently lack the full physiological context of living organisms, generating three-dimensional tissue-like structures in suspension poses additional challenges. Organoids remain limited in size—typically in the micrometer to low millimeter range—and differ functionally from native tissues, primarily due to the absence of a circulatory system. As their diameter increases (up to ~5 mm after months of culture), nutrient diffusion and waste removal become inefficient, leading to compromised viability of cells in the core [142]. In native tissues, vascular networks not only deliver oxygen and nutrients but also regulate organ function, homeostasis, and regeneration. Similarly, vascularization is critical in organoid cultures to support survival and functionality by facilitating exchange processes [143]. To address these limitations, microfluidic technologies are being integrated into 3D culture systems. These platforms enhance the physiological relevance of organoids by improving nutrient delivery, guiding growth, enabling real-time sensing, and supporting complex co-culture setups. Notably, self-assembling microfluidic devices allow antitumor testing to be conducted directly on the growth platform post spheroid formation, potentially reducing time and cost in optimizing drug regimens during preclinical evaluation. Advancements in 3D bioprinting and microfluidic technologies are also being investigated to develop tumor models that more accurately reflect the tumor microenvironment [144]. 3D bioprinting enables the precise placement of multiple cell types and ECM components, facilitating the formation of complex tissue-like structures that better reflect native tumor architecture. In parallel, microfluidic devices, often referred to as “tumor-on-a-chip” models, can simulate dynamic physiological conditions, such as fluid flow and mechanical forces, closely simulating the in vivo conditions [145,146,147,148]. These advancements enhance the physiological relevance of tumor models, offering greater insights into cancer biology and improving the predictive value of preclinical studies. Tumor-on-a-chip is a powerful device to investigate the EV-mediated crosstalk between tumor spheroids and surrounding healthy tissues, enabling the study of cancer progression, including cell invasion, angiogenesis, and metastasis [149,150].
By embedding them in extracellular matrix-like materials, it is possible to analyze how cancer cells invade surrounding tissues and, therefore, evaluate anti-metastatic agents [151,152]. This setup provides valuable insights into the molecular mechanisms driving cancer spread and opportunities for metastasis-involved pathway studies and, consequently, therapeutic intervention. Moreover, the 3D structure enriches cancer stem cell populations, facilitating studies on tumor initiation, progression, and recurrence, as well as therapies targeting these cells specifically [153,154,155].
Furthermore, tumor-on-a-chip systems are associated with a microfluidic system, making them a perfect platform for drug distribution and effectiveness in an environment that closely mimics in vivo conditions, providing reliable data for preclinical studies [156,157,158,159].
A key advantage of 3D models is their ability to predict the efficiency of anticancer drugs more accurately, as their 3D structure restricts drug penetration, simulating the barrier present in solid tumors involved in drug and immunotherapy resistance. Furthermore, they serve as an important platform for anticancer drug screening, enabling the evaluation of both the cytotoxicity and efficacy of novel therapeutic agents [124,160,161,162]. High-throughput screening methodologies further enhance their utility, allowing for the testing of large libraries of compounds on spheroids, and identifying potential anticancer drugs that might be overlooked using traditional monolayer assays [158,163,164,165].

6. EVs from Tumor Spheroids

Monolayer cultures have traditionally been widely used for isolating EVs (2D EVs) due to reproducible and well-established isolation methods as well as ease of manipulation. However, limitations of 2D systems have prompted a transition to 3D cultures, such as spheroids, which more accurately reflect in vivo conditions (Figure 4).
It is well documented that 3D cultures not only increase EV secretion but also alter EV composition, particularly by enriching their RNA cargo [166,167,168]. For example, mesenchymal stem cell (MSC) spheroids show increased EV production and altered RNA content compared to their 2D culture counterparts [169]. The cytokine secretion profile of MSC spheroids is influenced by their size: larger spheroids tend to release more pro-angiogenic and anti-inflammatory factors, while smaller spheroids mitigate cellular senescence, promoting the release of pro-angiogenic molecules. Additionally, 3D cultures also enhance the ability of MSCs to selectively release signaling factors through EVs, bolstering their immunomodulatory effects [170]. Kim and colleagues compared the cargo composition of EVs produced from 2D or 3D MSC culture, performing a comparative molecular profiling using proteomics and microRNA sequencing [171]. In this manner, they identified 224 miRNAs expressed in both systems, but 44 miRNAs expressed only in 2D EVs and 130 exclusive to 3D EVs. Among them, the immunomodulatory cytokine TGF-β1 and let-7b-5p miRNA, which negatively regulate the TLR4/NF-κB pathway, are amply upregulated in 3D systems. In gastric cancer, comparison between 2D and 3D systems showed upregulation of microRNA and downregulation of proteins involved in the ADP-ribosylation factor 6 (ARF6) signaling pathways in EVs produced by spheroids, highlighting the impact of cellular organization on EV biogenesis and molecular content [172].
This enhanced EV functionality has notable therapeutic implications. For instance, EVs derived from human dermal fibroblast spheroids exhibit anti-skin-aging properties on skin, suggesting their potential application in preventing and treating cutaneous aging [173]. Similarly, EVs from dermal papilla cell spheroids, essential for hair growth, are enriched in miR-218-5p, an miRNA that upregulates β-catenin signaling, thereby promoting hair follicle development [174]. In regenerative medicine, secretomes and EVs derived from lung spheroids and MSC spheroids have been investigated for their potential in treating lung injury and fibrosis, with lung spheroid-derived factors showing notable, promising repair capabilities [175].
In cancer research, EVs derived from tumor spheroids (3D EVs) are considered more physiologically relevant than those from 2D cultures (2D EVs). For example, a study on cervical cancer spheroids revealed that 3D EVs exhibit a small RNA profile remarkably similar (~96%) to that of in vivo circulating EVs found in the plasma of cervical cancer patients [176]. This high degree of similarity indicates a more accurate model for investigating tumor biomarkers, drug screening, and understanding the molecular mechanisms of tumor progression and metastasis.
3D cultures have shed light on the role of the tumor microenvironment in EV-mediated intercellular communication. For example, in ovarian cancer spheroids, exposure to cisplatin alters the miRNA cargo of secreted EVs, increasing the migration of bone marrow MSCs (BM-MSCs). These activated BM-MSCs subsequently increase secretion of interleukin-6 (IL-6), interleukin-8 (IL-8), and vascular endothelial growth factor A (VEGFA); promote angiogenesis in endothelial cells; and stimulate the migration of low-invasive ovarian cancer cells. Therefore, cisplatin can facilitate pro-tumorigenic behavior by modulating EV content [177,178]. In addition to secreted EVs, a subset of vesicles is trapped within the multicellular ovarian cancer spheroids known as “inner” EVs, participating in vasculogenic mimicry, a process in which cancer cells form vascular-like, tube-shaped structures. This process allows cancer cells to catch oxygen and nutrients and become independent from the presence of endothelial cells and traditional angiogenetic mechanisms [178]. Moreover, the role of EVs in pancreatic tumor-induced cachexia was investigated by targeting ZIP4, a zinc transporter implicated in tumor growth and metastasis. Silencing ZIP4 leads to reduced expression of the small GTPase RAB27B, which in turn decreases the release of heat shock proteins Hsp70 and Hsp90, two proteins commonly found in EVs [179].
3D cell culture systems have also been employed to investigate the role of the tumor microenvironment and its potential contribution to cancer EV secretion. A study using heterotypic melanoma cells and preadipocytes revealed an exosomal crosstalk between the two cell types that promotes cancer progression and metastasis through miR-155 expression [180]. Such findings may provide insights into using miRNA modulation as a therapeutic strategy to inhibit or treat melanoma. Similarly, EVs released from non-small-cell lung cancer (NSCLC) actively modulate the tumor microenvironment, supporting cancer proliferation and enhancing signaling to inhibit apoptosis, thereby promoting cancer metastasis [181]. Donzelli and coworkers also developed a 3D spheroid heterotypic culture model of NSCLC cell lines and fibroblasts to investigate the interplay between EVs, miR-574-5p, and the inflammatory mediator prostaglandin E2 (PGE2). Therefore, EVs containing miR-574-5p are taken up by neighboring cancer cells, triggering the upregulation of PGE2 synthesis [182]. This highlights the intricate nature of EV interactions and the advantages of using multicellular 3D culture models to clarify mechanisms arising from these complex interactions, potentially targeted for future therapeutic purposes [183]. Further evidence of the microenvironment’s impact on EV secretion has also been observed from the comparison between spheroids and xenograft models. In vitro, multicellular spheroids from patient-derived colorectal cancer secrete higher quantities of EVs, with release occurring from apical and basolateral cancer cell domains. In contrast, xenograft tumor spheroid models demonstrate EV release from all cancer cell domains, and the overall quantity of EVs is significantly greater compared to those released from spheroid cultures [184].
On the other hand, 3D systems also permit the investigation of the role of immune cells in cancer progression, because the interplay between cancer and immune cells via EVs within the TME plays a pivotal role in tumor development and modulation of immune responses [185]. Notably, differences in the composition and function of tumor-derived EV between 2D and 3D cell culture models can significantly impact the anti-cancer immune response. For instance, compared to traditional 2D glioblastoma cultures, extracellular vesicles released by 3D tumor organoids displayed a higher abundance of miRNAs involved in immunoregulatory signaling, such as interleukin-4 (IL-4) and interleukin-13 (IL-13) cytokine pathways. They are involved in tumor proliferation and in immunosuppressive phenotypes by inducing macrophages to polarize into an M2-like phenotype [186]. A difference in EV composition and effect between 2D and 3D systems was also observed in the breast cancer model. EVs from spheroids carried a higher number of pro-inflammatory and pro-tumorigenic molecules related to Natural Killer (NK) cell activation. This was evident when EVs derived from breast cancer spheroids were used to treat peripheral blood mononuclear cells (PBMCs) from healthy donors. This treatment led to both activation and reduction in the proportion of CD335+/CD11b+ Natural Killer (NK) cells, along with a significant decrease in CD39+ regulatory T cells (T-reg) involved in suppressing excessive immune responses related to the inflammatory response [187]. In addition, β-catenin-mutated tumors, which are known to exhibit resistance to immunotherapy, have been further investigated in the context of liver cancer. A recent study analyzing β-catenin activation in liver cancer cell lines and hepatocellular carcinoma patient samples revealed that activation of this pathway correlates with a reduction in exosome release and diminished immune cell infiltration [188].
Spheroids have proven to be invaluable tools for studying cancer stem cells (CSCs) and their role within tumor tissues. In a comprehensive study involving 67 cancer cell lines cultured under 3D conditions, spheroids were classified based on cellular aggregates formed and analyzed for gene expression profiles, with a focus on oncogenes and stem cell genes. Cells forming 3D structures were shown to efficiently secrete tumor EVs positive for epithelial cell adhesion molecule (EpCAM) and HSP90. These EVs were capable of transforming induced pluripotent cells into CSC-like cells, highlighting the functional role in maintaining CSC populations and driving tumor progression [189].
Additionally, cancer-derived EVs contain circular RNA (circRNAs) with significant regulatory roles [190]. A study using a breast cancer spheroid model demonstrated that EVs from cancer cells contain elevated levels of circRNAs compared to EVs from normal breast cells. Among this, a specific circRNA was found to regulate glycolysis in breast cancer cells via miR-1252-5p-mediated regulation of PFKFB2 (6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 2) expression, emphasizing the metabolic reprogramming potential of EVs from cancer cells [191].
Moreover, 3D culture systems have significantly advanced the study of EVs as potential cancer biomarkers for diagnosis, prognosis, and treatment monitoring [192,193]. Compared to nonneoplastic individuals, cancer patients exhibit a significantly higher concentration of EVs in body fluids, along with altered molecular composition. As a result, the miRNA and protein cargo of EVs can be investigated as promising biomarkers. Since 3D models more accurately simulate the in vivo tumor environment, EVs secreted from 3D cultures closely reflect the molecular signals observed in clinical settings. For instance, EVs derived from gastric cancer spheroids exhibit a general upregulation of microRNAs and a downregulation of protein content, indicating a shift in their molecular profile. These compositional changes influence both the biological activity of EVs and their uptake by recipient cells, making 3D cultures a more physiologically relevant and high-throughput alternative to traditional 2D in vitro systems [172]. Additionally, EVs released by pancreatic ductal adenocarcinoma (PDAC) tumor organoids have been compared with plasma EVs from patients with PDAC, benign gastrointestinal diseases, and chronic pancreatitis. This comparative analysis led to the identification of four EV proteins as potential novel biomarkers for PDAC, demonstrating the clinical utility of 3D culture-derived EVs in cancer diagnosis and prognosis [194].
Despite growing interest, significant challenges remain in translating tumor spheroid-derived extracellular vesicles (3D EVs) into clinically effective therapeutics. While several studies have reported good manufacturing practice (GMP)-compliant and scalable methods for producing 3D EVs, most of these rely on conventional 2D cultures, which limit scalability and physiological relevance [195]. In contrast, 3D micro-patterned well systems offer a promising alternative for large-scale EV production, with evidence showing improved therapeutic outcomes—such as enhanced morphology and connectivity—in preclinical stroke models. To address these limitations, Son and colleagues developed a bioprocessing platform based on a non-adhesive, microwell-patterned 3D culture system optimized for the serum-free production of EVs from human Wharton’s Jelly-derived MSCs (WJ-MSCs) and compared the yield, size distribution, and purity of EVs produced in this 3D system (3D EVs) to those derived from standard 2D MSC cultures (natural EVs), highlighting the advantages of the 3D approach for consistent and scalable EV manufacturing [196].

7. EVs from Tumor Organoids

Organoid-derived EVs present unique challenges and complexities not typically encountered with EVs from traditional 2D cell cultures. One major factor is the structural and cellular heterogeneity of organoids, which are composed of multiple cell types. This diversity leads to a wider array of EV subpopulations, each potentially varying in size, morphology, and biological function. Consequently, standard characterization tools like Transmission Electron Microscopy (TEM) must be interpreted with caution due to this added complexity. In addition to their biological intricacy, organoids require advanced culturing systems, usually involving three-dimensional matrices, that further complicate EV isolation. Components of these matrices, including residual proteins or gel fragments, can contaminate EV preparations and hinder downstream analysis. Isolating EVs from organoid systems demands highly refined techniques and greater precision than those used for standard 2D cell culture models to preserve vesicle purity and functionality. Nevertheless, EV studies using organoids have distinct benefits, one of which is the limitation of in vivo experiments using a model that closely resembles the native tissue, with a closer physiologic relevance over 2D culture.
The study of EVs from 3D in vitro culture systems mainly focuses on 3D spheroid tumor models, which have some characteristics that better represent the system, such as the necrotic core. However, organoid-derived EVs are emerging as critical mediators of cancer progression, intercellular signaling, and diagnostic potential, with studies across tumor types offering unique mechanistic insights (Table 1). In pancreatic cancer, particularly pancreatic ductal adenocarcinoma (PDAC), several studies emphasize EVs’ diagnostic and pathogenic relevance. A 2021 study showed that PDAC organoids recapitulate tumor heterogeneity and share EV miRNA signatures with matched patient plasma, including miR-21 and miR-195, while also revealing that extracellular matrix remodeling (e.g., collagen I deposition) boosts EV release in both PDAC and chronic pancreatitis, explaining elevated circulating EV levels in both conditions [197]. Moreover, the implementation, using a 3D biomimetic PDAC model that integrates tumor organoids and host-matching stromal cancer-associated fibroblasts (CAFs), showed that the matrix stiffness activates CAFs to increase exosome secretion, driving chemoresistance, a process reversible by exosome inhibitors like climbazole and imipramine [198]. PDAC with cachexia-related muscle wasting was investigated in 2025 through EVs as a mediator in PDAC–skeletal muscle communication. PDAC-derived EVs enriched in miR-223-5p promote muscle wasting through suppression of METTL14 via MAFA targeting, linking circulating EV miRNA content to cachexia and poor prognosis [199]. Researchers have also analyzed the protein profiles of EVs from PDAC organoids using mass spectrometry to distinguish EVs from PDAC and healthy pancreatic organoids. They observed that tumor-derived EVs are enriched in pro-tumorigenic proteins like LAMA5 and SDCBP, highlighting their utility for early diagnosis and monitoring [200]. In colorectal cancer (CRC), organoid-based studies consistently demonstrate the role of EVs in disease progression and biomarker development. An investigation focusing on the adenoma-to-carcinoma transition highlighted miR-1246 upregulation in CRC organoid-derived EVs, showing its role in promoting proliferation [201]. Another study identified APC, a common tumor suppressor gene, and its mutation is associated with familial adenomatous polyposis (FAP) and sporadic colorectal tumors. APC mutation and collagen deposition are critical enhancers of EV release in CRC organoids, with fibroblast-derived EVs shown to induce organoid colony formation under hypoxia, supporting a reciprocal tumor–stroma EV axis [202]. The researchers knocked out MMP3 in tumor organoids, highlighting MMP3’s dual role in tumoroid structure and EV integrity, showing that MMP3-rich EVs rescue proliferation and CD9/CD63 expression in deficient organoids, thereby positioning MMP3 as a key modulator of tumor EV function [203]. A 2025 multicenter study identified four exosomal miRNAs (miR-4284, miR-5100, miR-1246, miR-1290) elevated in CRC patient serum, showing diagnostic performance comparable to carcinoembryonic antigen (CEA) and improved accuracy when combined [204]. Earlier foundational work in 2013 on LIM1863 colon carcinoma organoids isolated two distinct EV subpopulations (apical and basolateral), with distinct proteomic profiles, including tumor-promoting complexes such as EpCAM, claudin-7, and CD44, demonstrating spatial and functional heterogeneity of EVs [205]. In glioblastoma, a 2024 study comparing 2D and 3D models found that organoid-derived EVs are enriched in immune-modulatory miRNAs and proteins, underscoring 3D models’ advantage in capturing glioblastoma EV biology [186]. Furthermore, studies across multiple cancer types deepen our understanding of EV regulation. An investigation across lung and pancreatic organoids revealed that Wnt signaling is tightly coupled to cell proliferation and EV secretion, with this link disrupted in PDAC but preserved in lung adenocarcinoma, emphasizing tissue-specific EV regulatory mechanisms [206].
Collectively, these studies demonstrate that organoid-derived EVs provide a robust platform for elucidating tumor biology, intercellular signaling, and biomarker discovery, with their context-specific cargo and behavior offering powerful avenues for precision oncology.

8. Conclusions

The transition from 2D to 3D cell culture systems represents a significant advancement in EV research, providing more physiologically relevant models that closely mimic the complexities of in vivo environments. While 2D monolayer cultures have been instrumental due to their reproducibility and ease of EV isolation, they fail to capture the intricate cellular interactions and microenvironmental dynamics of native tissues. In contrast, 3D cultures, such as spheroids and organoids, overcome these limitations, offering a closer mimicry of native tissue structures and enhancing the biological relevance of EV studies. Moreover, 3D culture techniques enable the growth of larger cell quantities in space- and cost-efficient ways, which are essential for the clinical application of EV-based therapies.
Spheroids derived from cancer cells showed increased EV production, with altered RNA content and cytokine profiles depending on spheroid size and composition. These characteristics have broadened the therapeutic potential of EVs for applications in tissue repair, immune modulation, and targeted therapy, as evidenced by their roles in lung injury and skin anti-aging studies. Notably, 3D-derived EVs from tumor spheroids more closely resemble those found in cancer patients, making them invaluable for studying tumor progression, metastasis, and therapeutic responses.
Organoid-derived EVs significantly deepen the landscape of EV research by more accurately recapitulating tissue-specific architecture and the complex multicellular interactions present in native tumors. This enables more precise investigation of tumor–stroma crosstalk, immune modulation, and mechanisms of drug resistance. These systems have enabled the discovery of cancer-specific EV cargo with strong potential as diagnostic and prognostic biomarkers. However, their biological complexity also introduces technical challenges in EV isolation and characterization, particularly due to matrix contamination and heterogeneity of vesicle populations.
In conclusion, EVs released by 3D tumor models, including spheroids and organoids, represent a promising area of study with significant implications for understanding tumor biology and developing novel cancer therapies. These vesicles not only provide a window into the complex intercellular communication within tumors but also offer potential as biomarkers and therapeutic targets. Future efforts should address the challenges in EV isolation, large-scale production, and characterization to facilitate their translation into clinical applications. By bridging the gap between in vitro and in vivo systems, 3D cultures enable more accurate modeling of disease processes and offer a powerful platform for developing targeted, effective cancer treatments.

Funding

This research was funded by the SiciliAn MicronanOTecH Research And Innovation CEnter “SAMOTHRACE” (MUR, PNRR-M4C2, ECS_00000022) and Spoke 3-Università degli Studi di Palermo “S2-COMMs-Micro and Nanotechnologies for Smart & Sustainable Communities”.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
2DTwo-dimensional
3DThree-dimensional
3D EVsTumor spheroid-derived extracellular vesicles
ARF6ADP-ribosylation factor 6
ASCsAdult tissue-resident stem cells
BMDCBone marrow-derived cells
CAFCancer-associated fibroblast
CEACarcinoembryonic antigen
CRCColorectal cancer
ECsEndothelial cells
ESCRTEndosomal Sorting Complex Required for Transport
ESCsEmbryonic stem cells
EVsExtracellular vesicles
FAPFamilial adenomatous polyposis
FBSFetal bovine serum
FcFragment crystallizable
FLT1Vascular endothelial growth factor receptor
GBMGlioblastoma
GelMAGelatin methacrylate
GMPGood manufacturing practice
HAHyaluronic acid
HSCHepatic stellate cell
IL-4Interleukin-4
IL-6Interleukin-6
IL-8Interleukin-8
IL-13Interleukin-13
ILVIntraluminal vesicle
iPSCsInduced pluripotent stem cells
MDSCMyeloid-derived suppressor cell
METTL3Methyltransferase-like 3
MIO NPsMagnetic iron oxide nanoparticles
MMPsMatrix metalloproteinases
MTSMulticellular tumor spheroids
MVBsMultivesicular bodies
MVsMicrovesicles
MSCsMesenchymal stromal cells
NGFNerve growth factor
NGFRNGF receptor
NKNatural Killer
PBMCsPeripheral blood mononuclear cells
PD-1Death protein 1
PDACPancreatic ductal adenocarcinoma
PD-L1Programmed death-ligand 1
PDOsPatient-derived organoids
pHEMAPoly(2-hydroxyethyl methacrylate)
PMNPre-metastatic niches
SNARESNAP receptor
SNAPSoluble NSF [N-ethylmaleimide-Sensitive Factor] attachment protein
TAMTumor-associated macrophages
TEMTransmission Electron Microscopy
TMETumor microenvironment
TMZTemozolomide
TNBCTriple-negative breast cancer
T-regRegulatory T cells
VEGFVascular endothelial growth factor
WJ-MSCsWharton’s Jelly-derived MSCs

References

  1. Raposo, G.; Stoorvogel, W. Extracellular Vesicles: Exosomes, Microvesicles, and Friends. J. Cell Biol. 2013, 200, 373–383. [Google Scholar] [CrossRef] [PubMed]
  2. van Niel, G.; Carter, D.R.F.; Clayton, A.; Lambert, D.W.; Raposo, G.; Vader, P. Challenges and Directions in Studying Cell–Cell Communication by Extracellular Vesicles. Nat. Rev. Mol. Cell Biol. 2022, 23, 369–382. [Google Scholar] [CrossRef] [PubMed]
  3. Lo Cicero, A.; Stahl, P.D.; Raposo, G. Extracellular Vesicles Shuffling Intercellular Messages: For Good or for Bad. Curr. Opin. Cell Biol. 2015, 35, 69–77. [Google Scholar] [CrossRef]
  4. Kalluri, R.; LeBleu, V.S. The Biology, Function, and Biomedical Applications of Exosomes. Science 2020, 367, eaau6977. [Google Scholar] [CrossRef]
  5. Riches, A.; Campbell, E.; Borger, E.; Powis, S. Regulation of Exosome Release from Mammary Epithelial and Breast Cancer Cells-A New Regulatory Pathway. Eur. J. Cancer 2014, 50, 1025–1034. [Google Scholar] [CrossRef]
  6. Kosaka, N.; Iguchi, H.; Hagiwara, K.; Yoshioka, Y.; Takeshita, F.; Ochiya, T. Neutral Sphingomyelinase 2 (NSMase2)-Dependent Exosomal Transfer of Angiogenic Micrornas Regulate Cancer Cell Metastasis. J. Biol. Chem. 2013, 288, 10849–10859. [Google Scholar] [CrossRef]
  7. Cook, K.; Li, H. Advancing Extracellular Vesicle Production: Improving Physiological Relevance and Yield with 3D Cell Culture. Nanoscale 2025, 17, 15110–15131. [Google Scholar] [CrossRef]
  8. Khan, N.L.A.; Muhandiram, S.; Dissanayake, K.; Godakumara, K.; Midekessa, G.; Andronowska, A.; Heath, P.R.; Kodithuwakku, S.; Hart, A.R.; Fazeli, A. Effect of 3D and 2D Cell Culture Systems on Trophoblast Extracellular Vesicle Physico-Chemical Characteristics and Potency. Front. Cell Dev. Biol. 2024, 12, 1382552. [Google Scholar] [CrossRef]
  9. Wu, B.; Liu, D.A.; Guan, L.; Myint, P.K.; Chin, L.K.; Dang, H.; Xu, Y.; Ren, J.; Li, T.; Yu, Z.; et al. Stiff Matrix Induces Exosome Secretion to Promote Tumour Growth. Nat. Cell Biol. 2023, 25, 415–424. [Google Scholar] [CrossRef]
  10. Paniushkina, L.; Grueso-Navarro, E.; Cheng, X.; Nazarenko, I. Three-Dimensional Cell Models for Extracellular Vesicles Production, Isolation, and Characterization. Methods Enzymol. 2020, 645, 209–230. [Google Scholar] [CrossRef]
  11. Abuwatfa, W.H.; Pitt, W.G.; Husseini, G.A. Scaffold-Based 3D Cell Culture Models in Cancer Research. J. Biomed. Sci. 2024, 31, 7. [Google Scholar] [CrossRef]
  12. Krylova, S.V.; Feng, D. The Machinery of Exosomes: Biogenesis, Release, and Uptake. Int. J. Mol. Sci. 2023, 24, 1337. [Google Scholar] [CrossRef] [PubMed]
  13. Vats, S.; Galli, T. Role of SNAREs in Unconventional Secretion—Focus on the VAMP7-Dependent Secretion. Front. Cell Dev. Biol. 2022, 10, 884020. [Google Scholar] [CrossRef] [PubMed]
  14. Colombo, M.; Raposo, G.; Théry, C. Biogenesis, Secretion, and Intercellular Interactions of Exosomes and Other Extracellular Vesicles. Annu. Rev. Cell Dev. Biol. 2014, 30, 255–289. [Google Scholar] [CrossRef] [PubMed]
  15. Ghossoub, R.; Lembo, F.; Rubio, A.; Gaillard, C.B.; Bouchet, J.; Vitale, N.; Slavík, J.; Machala, M.; Zimmermann, P. Syntenin-ALIX Exosome Biogenesis and Budding into Multivesicular Bodies Are Controlled by ARF6 and PLD2. Nat. Commun. 2014, 5, 3477. [Google Scholar] [CrossRef]
  16. Henne, W.M.; Stenmark, H.; Emr, S.D. Molecular Mechanisms of the Membrane Sculpting ESCRT Pathway. Cold Spring Harb. Perspect. Biol. 2013, 5, a016766. [Google Scholar] [CrossRef]
  17. Colombo, M.; Moita, C.; Van Niel, G.; Kowal, J.; Vigneron, J.; Benaroch, P.; Manel, N.; Moita, L.F.; Théry, C.; Raposo, G. Analysis of ESCRT Functions in Exosome Biogenesis, Composition and Secretion Highlights the Heterogeneity of Extracellular Vesicles. J. Cell Sci. 2013, 126, 5553–5565. [Google Scholar] [CrossRef]
  18. Lo Cicero, A.; Raposo, G. The Cell Biology of Exosomes: Historical and Perspectives. In Emerging Concepts of Tumor Exosome–Mediated Cell-Cell Communication; Springer: New York, NY, USA, 2012; pp. 1–32. [Google Scholar] [CrossRef]
  19. Trajkovic, K.; Hsu, C.; Chiantia, S.; Rajendran, L.; Wenzel, D.; Wieland, F.; Schwille, P.; Brügger, B.; Simons, M. Ceramide Triggers Budding of Exosome Vesicles into Multivesicular Endosomes. Science 2008, 319, 1244–1247. [Google Scholar] [CrossRef]
  20. Edgar, J.R.; Eden, E.R.; Futter, C.E. Hrs- and CD63-Dependent Competing Mechanisms Make Different Sized Endosomal Intraluminal Vesicles. Traffic 2014, 15, 197–211. [Google Scholar] [CrossRef]
  21. Palmulli, R.; Couty, M.; Piontek, M.C.; Ponnaiah, M.; Dingli, F.; Verweij, F.J.; Charrin, S.; Tantucci, M.; Sasidharan, S.; Rubinstein, E.; et al. CD63 Sorts Cholesterol into Endosomes for Storage and Distribution via Exosomes. Nat. Cell Biol. 2024, 26, 1093–1109. [Google Scholar] [CrossRef]
  22. Fan, Y.; Pionneau, C.; Cocozza, F.; Boëlle, P.Y.; Chardonnet, S.; Charrin, S.; Théry, C.; Zimmermann, P.; Rubinstein, E. Differential Proteomics Argues against a General Role for CD9, CD81 or CD63 in the Sorting of Proteins into Extracellular Vesicles. J. Extracell. Vesicles 2023, 12, e12352. [Google Scholar] [CrossRef]
  23. Ostenfeld, M.S.; Jeppesen, D.K.; Laurberg, J.R.; Boysen, A.T.; Bramsen, J.B.; Primdal-Bengtson, B.; Hendrix, A.; Lamy, P.; Dagnaes-Hansen, F.; Rasmussen, M.H.; et al. Cellular Disposal of MiR23b by RAB27-Dependent Exosome Release Is Linked to Acquisition of Metastatic Properties. Cancer Res. 2014, 74, 5758–5771. [Google Scholar] [CrossRef]
  24. Wang, G.; Li, J.; Bojmar, L.; Chen, H.; Li, Z.; Tobias, G.C.; Hu, M.; Homan, E.A.; Lucotti, S.; Zhao, F.; et al. Tumour Extracellular Vesicles and Particles Induce Liver Metabolic Dysfunction. Nature 2023, 618, 374–382. [Google Scholar] [CrossRef]
  25. Peche, V.S.; Pietka, T.A.; Jacome-Sosa, M.; Samovski, D.; Palacios, H.; Chatterjee-Basu, G.; Dudley, A.C.; Beatty, W.; Meyer, G.A.; Goldberg, I.J.; et al. Endothelial Cell CD36 Regulates Membrane Ceramide Formation, Exosome Fatty Acid Transfer and Circulating Fatty Acid Levels. Nat. Commun. 2023, 14, 4029. [Google Scholar] [CrossRef]
  26. Boshans, R.L.; Szanto, S.; van Aelst, L.; D’Souza-Schorey, C. ADP-Ribosylation Factor 6 Regulates Actin Cytoskeleton Remodeling in Coordination with Rac1 and RhoA. Mol. Cell. Biol. 2000, 20, 3685–3694. [Google Scholar] [CrossRef] [PubMed]
  27. Liu, L.; Kawashima, M.; Sugimoto, M.; Sonomura, K.; Pu, F.; Li, W.; Takeda, M.; Goto, T.; Kawaguchi, K.; Sato, T.A.; et al. Discovery of Lipid Profiles in Plasma-Derived Extracellular Vesicles as Biomarkers for Breast Cancer Diagnosis. Cancer Sci. 2023, 114, 4020–4031. [Google Scholar] [CrossRef] [PubMed]
  28. Skotland, T.; Hessvik, N.P.; Sandvig, K.; Llorente, A. Exosomal Lipid Composition and the Role of Ether Lipids and Phosphoinositides in Exosome Biology. J. Lipid Res. 2019, 60, 9–18. [Google Scholar] [CrossRef] [PubMed]
  29. Pfrieger, F.W.; Vitale, N. Thematic Review Series: Exosomes and Microvesicles: Lipids as Key Components of Their Biogenesis and Functions Cholesterol and the Journey of Extracellular Vesicles. J. Lipid Res. 2018, 59, 2255–2261. [Google Scholar] [CrossRef]
  30. Lauwers, E.; Wang, Y.C.; Gallardo, R.; Van der Kant, R.; Michiels, E.; Swerts, J.; Baatsen, P.; Zaiter, S.S.; McAlpine, S.R.; Gounko, N.V.; et al. Hsp90 Mediates Membrane Deformation and Exosome Release. Mol. Cell 2018, 71, 689–702.e9. [Google Scholar] [CrossRef]
  31. Nolte’T Hoen, E.N.M.; Buermans, H.P.J.; Waasdorp, M.; Stoorvogel, W.; Wauben, M.H.M.; ’T Hoen, P.A.C. Deep Sequencing of RNA from Immune Cell-Derived Vesicles Uncovers the Selective Incorporation of Small Non-Coding RNA Biotypes with Potential Regulatory Functions. Nucleic Acids Res. 2012, 40, 9272–9285. [Google Scholar] [CrossRef]
  32. Skog, J.; Würdinger, T.; van Rijn, S.; Meijer, D.H.; Gainche, L.; Curry, W.T.; Carter, B.S.; Krichevsky, A.M.; Breakefield, X.O. Glioblastoma Microvesicles Transport RNA and Proteins That Promote Tumour Growth and Provide Diagnostic Biomarkers. Nat. Cell Biol. 2008, 10, 1470–1476. [Google Scholar] [CrossRef]
  33. Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.F.; Li, S.; Chin, A.R.; et al. Breast-Cancer-Secreted MiR-122 Reprograms Glucose Metabolism in Premetastatic Niche to Promote Metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef]
  34. Sruthi, T.V.; Edatt, L.; Raji, G.R.; Kunhiraman, H.; Shankar, S.S.; Shankar, V.; Ramachandran, V.; Poyyakkara, A.; Kumar, S.V.B. Horizontal Transfer of MiR-23a from Hypoxic Tumor Cell Colonies Can Induce Angiogenesis. J. Cell. Physiol. 2018, 233, 3498–3514. [Google Scholar] [CrossRef]
  35. Kulkarni, B.; Gondaliya, P.; Kirave, P.; Rawal, R.; Jain, A.; Garg, R.; Kalia, K. Exosome-Mediated Delivery of MiR-30a Sensitize Cisplatin-Resistant Variant of Oral Squamous Carcinoma Cells via Modulating Beclin1 and Bcl2. Oncotarget 2020, 11, 1832–1845. [Google Scholar] [CrossRef] [PubMed]
  36. Cicero, A.L.; Delevoye, C.; Gilles-Marsens, F.; Loew, D.; Dingli, F.; Guéré, C.; André, N.; Vié, K.; Van Niel, G.; Raposo, G. Exosomes Released by Keratinocytes Modulate Melanocyte Pigmentation. Nat. Commun. 2015, 6, 7506. [Google Scholar] [CrossRef]
  37. Li, B.; Cao, Y.; Sun, M.; Feng, H. Expression, Regulation, and Function of Exosome-Derived MiRNAs in Cancer Progression and Therapy. FASEB J. 2021, e21916. [Google Scholar] [CrossRef] [PubMed]
  38. Xu, Z.; Chen, Y.; Ma, L.; Chen, Y.; Liu, J.; Guo, Y.; Yu, T.; Zhang, L.; Zhu, L.; Shu, Y. Role of Exosomal Non-Coding RNAs from Tumor Cells and Tumor-Associated Macrophages in the Tumor Microenvironment. Mol. Ther. 2022, 30, 3133–3154. [Google Scholar] [CrossRef]
  39. Dou, Q.; Wang, J.; Yang, Y.; Zhuo, W. Roles of Exosome-Derived Non-Coding RNA in Tumor Micro-Environment and Its Clinical Application. J. Zhejiang Univ. (Med. Sci.) 2025, 52, 429–438. [Google Scholar] [CrossRef] [PubMed]
  40. Waldenström, A.; Gennebäck, N.; Hellman, U.; Ronquist, G. Cardiomyocyte Microvesicles Contain DNA/RNA and Convey Biological Messages to Target Cells. PLoS ONE 2012, 7, e34653. [Google Scholar] [CrossRef]
  41. Fernando, M.R.; Jiang, C.; Krzyzanowski, G.D.; Ryan, W.L. New Evidence That a Large Proportion of Human Blood Plasma Cell-Free DNA Is Localized in Exosomes. PLoS ONE 2017, 12, e0183915. [Google Scholar] [CrossRef]
  42. Sansone, P.; Savini, C.; Kurelac, I.; Chang, Q.; Amato, L.B.; Strillacci, A.; Stepanova, A.; Iommarini, L.; Mastroleo, C.; Daly, L.; et al. Packaging and Transfer of Mitochondrial DNA via Exosomes Regulate Escape from Dormancy in Hormonal Therapy-Resistant Breast Cancer. Proc. Natl. Acad. Sci. USA 2017, 114, E9066–E9075. [Google Scholar] [CrossRef]
  43. Konaka, H.; Kato, Y.; Hirano, T.; Tsujimoto, K.; Park, J.; Koba, T.; Aoki, W.; Matsuzaki, Y.; Taki, M.; Koyama, S.; et al. Secretion of Mitochondrial DNA via Exosomes Promotes Inflammation in Behçet’s Syndrome. EMBO J. 2023, 42, e112573. [Google Scholar] [CrossRef] [PubMed]
  44. D’Acunzo, P.; Kim, Y.; Ungania, J.M.; Pérez-González, R.; Goulbourne, C.N.; Levy, E. Isolation of Mitochondria-Derived Mitovesicles and Subpopulations of Microvesicles and Exosomes from Brain Tissues. Nat. Protoc. 2022, 17, 2517–2549. [Google Scholar] [CrossRef] [PubMed]
  45. Xu, R.; Rai, A.; Chen, M.; Suwakulsiri, W.; Greening, D.W.; Simpson, R.J. Extracellular Vesicles in Cancer—Implications for Future Improvements in Cancer Care. Nat. Rev. Clin. Oncol. 2018, 15, 617–638. [Google Scholar] [CrossRef] [PubMed]
  46. Sriwastva, M.K.; Teng, Y.; Mu, J.; Xu, F.; Kumar, A.; Sundaram, K.; Malhotra, R.K.; Xu, Q.; Hood, J.L.; Zhang, L.; et al. An Extracellular Vesicular Mutant KRAS-Associated Protein Complex Promotes Lung Inflammation and Tumor Growth. J. Extracell. Vesicles 2023, 12, 12307. [Google Scholar] [CrossRef]
  47. Bhatta, B.; Luz, I.; Krueger, C.; Teo, F.X.; Lane, D.P.; Sabapathy, K.; Cooks, T. Cancer Cells Shuttle Extracellular Vesicles Containing Oncogenic Mutant P53 Proteins to the Tumor Microenvironment. Cancers 2021, 13, 2985. [Google Scholar] [CrossRef]
  48. Janpipatkul, K.; Panvongsa, W.; Worakitchanon, W.; Reungwetwattana, T.; Chairoungdua, A. Extracellular Vesicles from EGFR T790M/L858R-Mutant Non-Small Cell Lung Cancer Promote Cancer Progression. Anticancer. Res. 2022, 42, 3835–3844. [Google Scholar] [CrossRef]
  49. Li, Y.; Zheng, Y.; Tan, X.; Du, Y.; Wei, Y.; Liu, S. Extracellular Vesicle-Mediated Pre-Metastatic Niche Formation via Altering Host Microenvironments. Front. Immunol. 2024, 15, 1367373. [Google Scholar] [CrossRef]
  50. Li, M.X.; Hu, S.; Lei, H.H.; Yuan, M.; Li, X.; Hou, W.K.; Huang, X.J.; Xiao, B.W.; Yu, T.X.; Zhang, X.H.; et al. Tumor-Derived MiR-9-5p-Loaded EVs Regulate Cholesterol Homeostasis to Promote Breast Cancer Liver Metastasis in Mice. Nat. Commun. 2024, 15, 10539. [Google Scholar] [CrossRef]
  51. Cariello, M.; Squilla, A.; Piacente, M.; Venutolo, G.; Fasano, A. Drug Resistance: The Role of Exosomal MiRNA in the Microenvironment of Hematopoietic Tumors. Molecules 2022, 28, 116. [Google Scholar] [CrossRef]
  52. Peixoto da Silva, S.; Caires, H.R.; Bergantim, R.; Guimarães, J.E.; Vasconcelos, M.H. MiRNAs Mediated Drug Resistance in Hematological Malignancies. Semin. Cancer Biol. 2022, 83, 283–302. [Google Scholar] [CrossRef]
  53. Galati, D.; Solimando, A.G.; Reale, A.; Khong, T.; Spencer, A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. J. Clin. Med. 2022, 11, 6892. [Google Scholar] [CrossRef] [PubMed]
  54. Brennan, K.; Martin, K.; FitzGerald, S.P.; O’Sullivan, J.; Wu, Y.; Blanco, A.; Richardson, C.; Mc Gee, M.M. A Comparison of Methods for the Isolation and Separation of Extracellular Vesicles from Protein and Lipid Particles in Human Serum. Sci. Rep. 2020, 10, 1039. [Google Scholar] [CrossRef] [PubMed]
  55. Zhang, Q.; Jeppesen, D.K.; Higginbotham, J.N.; Franklin, J.L.; Coffey, R.J. Comprehensive Isolation of Extracellular Vesicles and Nanoparticles. Nat. Protoc. 2023, 18, 1462–1487. [Google Scholar] [CrossRef] [PubMed]
  56. Van Deun, J.; Mestdagh, P.; Agostinis, P.; Akay, Ö.; Anand, S.; Anckaert, J.; Martinez, Z.A.; Baetens, T.; Beghein, E.; Bertier, L.; et al. EV-TRACK: Transparent Reporting and Centralizing Knowledge in Extracellular Vesicle Research. Nat. Methods 2017, 14, 228–232. [Google Scholar] [CrossRef]
  57. Théry, C.; Witwer, K.W.; Aikawa, E.; Alcaraz, M.J.; Anderson, J.D.; Andriantsitohaina, R.; Antoniou, A.; Arab, T.; Archer, F.; Atkin-Smith, G.K.; et al. Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018): A Position Statement of the International Society for Extracellular Vesicles and Update of the MISEV2014 Guidelines. J. Extracell. Vesicles 2018, 7, 1535750. [Google Scholar] [CrossRef]
  58. Welsh, J.A.; Goberdhan, D.C.I.; O’Driscoll, L.; Buzas, E.I.; Blenkiron, C.; Bussolati, B.; Cai, H.; Di Vizio, D.; Driedonks, T.A.P.; Erdbrügger, U.; et al. Minimal Information for Studies of Extracellular Vesicles (MISEV2023): From Basic to Advanced Approaches. J. Extracell. Vesicles 2024, 13, e12404. [Google Scholar] [CrossRef]
  59. Greening, D.W.; Xu, R.; Ji, H.; Tauro, B.J.; Simpson, R.J. A Protocol for Exosome Isolation and Characterization: Evaluation of Ultracentrifugation, Density-Gradient Separation, and Immunoaffinity Capture Methods. Methods Mol. Biol. 2015, 1295, 179–209. [Google Scholar] [CrossRef]
  60. Bobrie, A.; Colombo, M.; Krumeich, S.; Raposo, G.; Théry, C. Diverse Subpopulations of Vesicles Secreted by Different Intracellular Mechanisms Are Present in Exosome Preparations Obtained by Differential Ultracentrifugation. J. Extracell. Vesicles 2012, 1, 18397. [Google Scholar] [CrossRef]
  61. Gupta, S.; Rawat, S.; Arora, V.; Kottarath, S.K.; Dinda, A.K.; Vaishnav, P.K.; Nayak, B.; Mohanty, S. An Improvised One-Step Sucrose Cushion Ultracentrifugation Method for Exosome Isolation from Culture Supernatants of Mesenchymal Stem Cells. Stem Cell Res. Ther. 2018, 9, 180. [Google Scholar] [CrossRef]
  62. Filipović, L.; Spasojević, M.; Prodanović, R.; Korać, A.; Matijaševic, S.; Brajušković, G.; de Marco, A.; Popović, M. Affinity-Based Isolation of Extracellular Vesicles by Means of Single-Domain Antibodies Bound to Macroporous Methacrylate-Based Copolymer. New Biotechnol. 2022, 69, 36–48. [Google Scholar] [CrossRef] [PubMed]
  63. Stranska, R.; Gysbrechts, L.; Wouters, J.; Vermeersch, P.; Bloch, K.; Dierickx, D.; Andrei, G.; Snoeck, R. Comparison of Membrane Affinity-Based Method with Size-Exclusion Chromatography for Isolation of Exosome-like Vesicles from Human Plasma. J. Transl. Med. 2018, 16, 1. [Google Scholar] [CrossRef] [PubMed]
  64. Filipe, V.; Hawe, A.; Jiskoot, W. Critical Evaluation of Nanoparticle Tracking Analysis (NTA) by NanoSight for the Measurement of Nanoparticles and Protein Aggregates. Pharm. Res. 2010, 27, 796–810. [Google Scholar] [CrossRef] [PubMed]
  65. Raposo, G.; Nijman, H.W.; Stoorvogel, W.; Leijendekker, R.; Harding, C.V.; Melief, C.J.M.; Geuze, H.J. B Lymphocytes Secrete Antigen-Presenting Vesicles. J. Exp. Med. 1996, 183, 1161–1172. [Google Scholar] [CrossRef]
  66. Pascucci, L.; Scattini, G. Imaging Extracelluar Vesicles by Transmission Electron Microscopy: Coping with Technical Hurdles and Morphological Interpretation. Biochim. Biophys. Acta Gen. Subj. 2021, 1865, 129648. [Google Scholar] [CrossRef]
  67. Nolan, J.P.; Duggan, E. Analysis of Individual Extracellular Vesicles by Flow Cytometry. Methods Mol. Biol. 2018, 1678, 79–92. [Google Scholar] [CrossRef]
  68. van der Vlist, E.J.; Nolte-’t Hoen, E.N.M.; Stoorvogel, W.; Arkesteijn, G.J.A.; Wauben, M.H.M. Fluorescent Labeling of Nano-Sized Vesicles Released by Cells and Subsequent Quantitative and Qualitative Analysis by High-Resolution Flow Cytometry. Nat. Protoc. 2012, 7, 1311–1326. [Google Scholar] [CrossRef]
  69. Pocsfalvi, G.; Stanly, C.; Vilasi, A.; Fiume, I.; Capasso, G.; Turiák, L.; Buzas, E.I.; Vékey, K. Mass Spectrometry of Extracellular Vesicles. Mass. Spectrom. Rev. 2016, 35, 3–21. [Google Scholar] [CrossRef]
  70. Acland, M.; Mittal, P.; Lokman, N.A.; Klingler-Hoffmann, M.; Oehler, M.K.; Hoffmann, P. Mass Spectrometry Analyses of Multicellular Tumor Spheroids. Proteom. Clin. Appl. 2018, 12, e1700124. [Google Scholar] [CrossRef]
  71. Ma, L.; Liu, Y.H.; Liu, C.; Wang, S.Q.; Ma, J.; Li, X.Q.; Ren, M.; Yang, T.T.; Liu, G.Z. LncRNA, MiRNA, and MRNA of Plasma and Tumor-Derived Exosomes of Cardiac Myxoma-Related Ischaemic Stroke. Sci. Data 2025, 12, 91. [Google Scholar] [CrossRef]
  72. Yuana, Y.; Sturk, A.; Nieuwland, R. Extracellular Vesicles in Physiological and Pathological Conditions. Blood Rev. 2013, 27, 31–39. [Google Scholar] [CrossRef]
  73. Chang, W.H.; Cerione, R.A.; Antonyak, M.A. Extracellular Vesicles and Their Roles in Cancer Progression. Methods Mol. Biol. 2021, 2174, 143–170. [Google Scholar] [CrossRef] [PubMed]
  74. Lo Cicero, A.; Majkowska, I.; Nagase, H.; Di Liegro, I.; Troeberg, L. Microvesicles Shed by Oligodendroglioma Cells and Rheumatoid Synovial Fibroblasts Contain Aggrecanase Activity. Matrix Biol. 2012, 31, 229–233. [Google Scholar] [CrossRef] [PubMed]
  75. Wandrey, M.; Jablonska, J.; Stauber, R.H.; Gül, D. Exosomes in Cancer Progression and Therapy Resistance: Molecular Insights and Therapeutic Opportunities. Life 2023, 13, 2033. [Google Scholar] [CrossRef] [PubMed]
  76. Minciacchi, V.R.; Freeman, M.R.; Di Vizio, D. Extracellular Vesicles in Cancer: Exosomes, Microvesicles and the Emerging Role of Large Oncosomes. Semin. Cell Dev. Biol. 2015, 40, 41–51. [Google Scholar] [CrossRef]
  77. Tang, M.K.S.; Yue, P.Y.K.; Ip, P.P.; Huang, R.L.; Lai, H.C.; Cheung, A.N.Y.; Tse, K.Y.; Ngan, H.Y.S.; Wong, A.S.T. Soluble E-Cadherin Promotes Tumor Angiogenesis and Localizes to Exosome Surface. Nat. Commun. 2018, 9, e1700124. [Google Scholar] [CrossRef]
  78. Chen, S.; Chen, X.; Luo, Q.; Liu, X.; Wang, X.; Cui, Z.; He, A.; He, S.; Jiang, Z.; Wu, N.; et al. Retinoblastoma Cell-Derived Exosomes Promote Angiogenesis of Human Vesicle Endothelial Cells through MicroRNA-92a-3p. Cell Death Dis. 2021, 12, 695. [Google Scholar] [CrossRef]
  79. Wang, L.; Qiao, C.; Han, L.; Wang, X.; Miao, J.; Cao, L.; Huang, C.; Wang, J. HOXD3 Promotes the Migration and Angiogenesis of Hepatocellular Carcinoma via Modifying Hepatocellular Carcinoma Cells Exosome-Delivered CCR6 and Regulating Chromatin Conformation of CCL20. Cell Death Dis. 2024, 15, 221. [Google Scholar] [CrossRef]
  80. Zhang, L.; Yu, D. Exosomes in Cancer Development, Metastasis, and Immunity. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 455–468. [Google Scholar] [CrossRef]
  81. Hoshino, A.; Costa-Silva, B.; Shen, T.L.; Rodrigues, G.; Hashimoto, A.; Tesic Mark, M.; Molina, H.; Kohsaka, S.; Di Giannatale, A.; Ceder, S.; et al. Tumour Exosome Integrins Determine Organotropic Metastasis. Nature 2015, 527, 329–335. [Google Scholar] [CrossRef]
  82. Taverna, S.; Flugy, A.; Saieva, L.; Kohn, E.C.; Santoro, A.; Meraviglia, S.; De Leo, G.; Alessandro, R. Role of Exosomes Released by Chronic Myelogenous Leukemia Cells in Angiogenesis. Int. J. Cancer 2012, 130, 2033–2043. [Google Scholar] [CrossRef]
  83. Wang, Y.; Jia, J.; Wang, F.; Fang, Y.; Yang, Y.; Zhou, Q.; Yuan, W.; Gu, X.; Hu, J.; Yang, S. Pre-Metastatic Niche: Formation, Characteristics and Therapeutic Implication. Signal Transduct. Target. Ther. 2024, 9, 236. [Google Scholar] [CrossRef] [PubMed]
  84. Peinado, H.; Alečković, M.; Lavotshkin, S.; Matei, I.; Costa-Silva, B.; Moreno-Bueno, G.; Hergueta-Redondo, M.; Williams, C.; García-Santos, G.; Ghajar, C.M.; et al. Melanoma Exosomes Educate Bone Marrow Progenitor Cells toward a Pro-Metastatic Phenotype through MET. Nat. Med. 2012, 18, 883–891. [Google Scholar] [CrossRef] [PubMed]
  85. García-Silva, S.; Benito-Martín, A.; Nogués, L.; Hernández-Barranco, A.; Mazariegos, M.S.; Santos, V.; Hergueta-Redondo, M.; Ximénez-Embún, P.; Kataru, R.P.; Lopez, A.A.; et al. Melanoma-Derived Small Extracellular Vesicles Induce Lymphangiogenesis and Metastasis through an NGFR-Dependent Mechanism. Nat. Cancer 2021, 2, 1387–1405. [Google Scholar] [CrossRef] [PubMed]
  86. Siddhartha, R.; Garg, M. Interplay Between Extracellular Matrix Remodeling and Angiogenesis in Tumor Ecosystem. Mol. Cancer Ther. 2023, 22, 291–305. [Google Scholar] [CrossRef]
  87. Kong, J.; Tian, H.; Zhang, F.; Zhang, Z.; Li, J.; Liu, X.; Li, X.; Liu, J.; Li, X.; Jin, D.; et al. Extracellular Vesicles of Carcinoma-Associated Fibroblasts Creates a Pre-Metastatic Niche in the Lung through Activating Fibroblasts. Mol. Cancer 2019, 18, 175. [Google Scholar] [CrossRef]
  88. González-Callejo, P.; Gener, P.; Díaz-Riascos, Z.V.; Conti, S.; Cámara-Sánchez, P.; Riera, R.; Mancilla, S.; García-Gabilondo, M.; Peg, V.; Arango, D.; et al. Extracellular Vesicles Secreted by Triple-Negative Breast Cancer Stem Cells Trigger Premetastatic Niche Remodeling and Metastatic Growth in the Lungs. Int. J. Cancer 2023, 152, 2153–2165. [Google Scholar] [CrossRef]
  89. Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic Cancer Exosomes Initiate Pre-Metastatic Niche Formation in the Liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
  90. Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 Contributes to Immunosuppression and Is Associated with Anti-PD-1 Response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef]
  91. Zhu, G.; Cao, B.; Liang, X.; Li, L.; Hao, Y.; Meng, W.; He, C.; Wang, L.; Li, L. Small Extracellular Vesicles Containing MiR-192/215 Mediate Hypoxia-Induced Cancer-Associated Fibroblast Development in Head and Neck Squamous Cell Carcinoma. Cancer Lett. 2021, 506, 11–22. [Google Scholar] [CrossRef]
  92. Li, Y.; Gao, S.; Hu, Q.; Wu, F. Functional Properties of Cancer Epithelium and Stroma-Derived Exosomes in Head and Neck Squamous Cell Carcinoma. Life 2022, 12, 757. [Google Scholar] [CrossRef] [PubMed]
  93. Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Twyman-Saint Victor, C.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome Transfer from Stromal to Breast Cancer Cells Regulates Therapy Resistance Pathways. Cell 2014, 159, 499–513. [Google Scholar] [CrossRef] [PubMed]
  94. Yang, X.; Zhang, Y.; Zhang, Y.; Li, H.; Li, L.; Wu, Y.; Chen, X.; Qiu, L.; Han, J.; Wang, Z. Colorectal Cancer-Derived Extracellular Vesicles Induce Liver Premetastatic Immunosuppressive Niche Formation to Promote Tumor Early Liver Metastasis. Signal Transduct. Target. Ther. 2023, 8, 102. [Google Scholar] [CrossRef] [PubMed]
  95. Nittayaboon, K.; Leetanaporn, K.; Sangkhathat, S.; Roytrakul, S.; Navakanitworakul, R. Proteomic Analysis of Butyrate-Resistant Colorectal Cancer-Derived Exosomes Reveals Potential Resistance to Anti-Cancer Drugs. Discov. Med. 2024, 36, 1306. [Google Scholar] [CrossRef] [PubMed]
  96. Andrade, L.N.d.S.; Otake, A.H.; Cardim, S.G.B.; da Silva, F.I.; Ikoma Sakamoto, M.M.; Furuya, T.K.; Uno, M.; Pasini, F.S.; Chammas, R. Extracellular Vesicles Shedding Promotes Melanoma Growth in Response to Chemotherapy. Sci. Rep. 2019, 9, 14482. [Google Scholar] [CrossRef]
  97. Yang, E.; Wang, L.; Jin, W.; Liu, X.; Wang, Q.; Wu, Y.; Tan, Y.; Wang, Y.; Cui, X.; Zhao, J.; et al. PTRF/Cavin-1 Enhances Chemo-Resistance and Promotes Temozolomide Efflux through Extracellular Vesicles in Glioblastoma. Theranostics 2022, 12, 4330–4347. [Google Scholar] [CrossRef]
  98. Serratì, S.; Guida, M.; Di Fonte, R.; De Summa, S.; Strippoli, S.; Iacobazzi, R.M.; Quarta, A.; De Risi, I.; Guida, G.; Paradiso, A.; et al. Circulating Extracellular Vesicles Expressing PD1 and PD-L1 Predict Response and Mediate Resistance to Checkpoint Inhibitors Immunotherapy in Metastatic Melanoma. Mol. Cancer 2022, 21, 20. [Google Scholar] [CrossRef]
  99. Yu, Z.L.; Liu, J.Y.; Chen, G. Small Extracellular Vesicle PD-L1 in Cancer: The Knowns and Unknowns. npj Precis. Oncol. 2022, 6, 42. [Google Scholar] [CrossRef]
  100. Kang, S.H.; Oh, S.Y.; Lee, K.Y.; Lee, H.J.; Kim, M.S.; Kwon, T.G.; Kim, J.W.; Lee, S.T.; Choi, S.Y.; Hong, S.H. Differential Effect of Cancer-Associated Fibroblast-Derived Extracellular Vesicles on Cisplatin Resistance in Oral Squamous Cell Carcinoma via MiR-876-3p. Theranostics 2024, 14, 460–479. [Google Scholar] [CrossRef]
  101. Chen, C.; Zhang, L.; Ruan, Z. GATA3 Encapsulated by Tumor-Associated Macrophage-Derived Extracellular Vesicles Promotes Immune Escape and Chemotherapy Resistance of Ovarian Cancer Cells by Upregulating the CD24/Siglec-10 Axis. Mol. Pharm. 2023, 20, 971–986. [Google Scholar] [CrossRef]
  102. Ning, J.; Hou, X.; Hao, J.; Zhang, W.; Shi, Y.; Huang, Y.; Ruan, X.; Zheng, X.; Gao, M. METTL3 Inhibition Induced by M2 Macrophage-Derived Extracellular Vesicles Drives Anti-PD-1 Therapy Resistance via M6A-CD70-Mediated Immune Suppression in Thyroid Cancer. Cell Death Differ. 2023, 30, 2265–2279. [Google Scholar] [CrossRef]
  103. Wiklander, O.P.B.; Mamand, D.R.; Mohammad, D.K.; Zheng, W.; Jawad Wiklander, R.; Sych, T.; Zickler, A.M.; Liang, X.; Sharma, H.; Lavado, A.; et al. Antibody-Displaying Extracellular Vesicles for Targeted Cancer Therapy. Nat. Biomed. Eng. 2024, 8, 1453–1468. [Google Scholar] [CrossRef] [PubMed]
  104. Bi, Y.; Chen, J.; Li, Q.; Li, Y.; Zhang, L.; Zhida, L.; Yuan, F.; Zhang, R. Tumor-Derived Extracellular Vesicle Drug Delivery System for Chemo-Photothermal-Immune Combination Cancer Treatment. iScience 2024, 27, 108833. [Google Scholar] [CrossRef] [PubMed]
  105. Hosseini, K.; Ranjbar, M.; Pirpour Tazehkand, A.; Asgharian, P.; Montazersaheb, S.; Tarhriz, V.; Ghasemnejad, T. Evaluation of Exosomal Non-Coding RNAs in Cancer Using High-Throughput Sequencing. J. Transl. Med. 2022, 20, 30. [Google Scholar] [CrossRef]
  106. Li, L.; Zhang, L.; Montgomery, K.C.; Jiang, L.; Lyon, C.J.; Hu, T.Y. Advanced Technologies for Molecular Diagnosis of Cancer: State of Pre-Clinical Tumor-Derived Exosome Liquid Biopsies. Mater. Today Bio 2023, 18, 100538. [Google Scholar] [CrossRef]
  107. Zhang, Z.; Liu, X.; Yang, X.; Jiang, Y.; Li, A.; Cong, J.; Li, Y.; Xie, Q.; Xu, C.; Liu, D. Identification of Faecal Extracellular Vesicles as Novel Biomarkers for the Non-Invasive Diagnosis and Prognosis of Colorectal Cancer. J. Extracell. Vesicles 2023, 12, 12300. [Google Scholar] [CrossRef]
  108. Yin, H.; Xie, J.; Xing, S.; Lu, X.; Yu, Y.; Ren, Y.; Tao, J.; He, G.; Zhang, L.; Yuan, X.; et al. Machine Learning-Based Analysis Identifies and Validates Serum Exosomal Proteomic Signatures for the Diagnosis of Colorectal Cancer. Cell Rep. Med. 2024, 5, 101689. [Google Scholar] [CrossRef]
  109. Yousafzai, N.A.; El Khalki, L.; Wang, W.; Szpendyk, J.; Sossey-Alaoui, K. Advances in 3D Culture Models to Study Exosomes in Triple-Negative Breast Cancer. Cancers 2024, 16, 883. [Google Scholar] [CrossRef]
  110. Rodrigues, D.B.; Reis, R.L.; Pirraco, R.P. Modelling the Complex Nature of the Tumor Microenvironment: 3D Tumor Spheroids as an Evolving Tool. J. Biomed. Sci. 2024, 31, 13. [Google Scholar] [CrossRef]
  111. Plava, J.; Cehakova, M.; Kuniakova, M.; Trnkova, L.; Cihova, M.; Bohac, M.; Danisovic, L. The Third Dimension of Tumor Microenvironment—The Importance of Tumor Stroma in 3D Cancer Models. Exp. Biol. Med. 2023, 248, 1347–1358. [Google Scholar] [CrossRef]
  112. El Harane, S.; Zidi, B.; El Harane, N.; Krause, K.H.; Matthes, T.; Preynat-Seauve, O. Cancer Spheroids and Organoids as Novel Tools for Research and Therapy: State of the Art and Challenges to Guide Precision Medicine. Cells 2023, 12, 1001. [Google Scholar] [CrossRef]
  113. Kirsh, S.M.; Pascetta, S.A.; Uniacke, J. Spheroids as a 3D Model of the Hypoxic Tumor Microenvironment. Methods Mol. Biol. 2023, 2614, 273–285. [Google Scholar] [CrossRef] [PubMed]
  114. Laurent, J.; Frongia, C.; Cazales, M.; Mondesert, O.; Ducommun, B.; Lobjois, V. Multicellular Tumor Spheroid Models to Explore Cell Cycle Checkpoints in 3D. BMC Cancer 2013, 13, 73. [Google Scholar] [CrossRef] [PubMed]
  115. Riffle, S.; Pandey, R.N.; Albert, M.; Hegde, R.S. Linking Hypoxia, DNA Damage and Proliferation in Multicellular Tumor Spheroids. BMC Cancer 2017, 17, 338. [Google Scholar] [CrossRef] [PubMed]
  116. Bhattacharya, S.; Calar, K.; De La Puente, P. Mimicking Tumor Hypoxia and Tumor-Immune Interactions Employing Three-Dimensional in Vitro Models. J. Exp. Clin. Cancer Res. 2020, 39, 75. [Google Scholar] [CrossRef]
  117. Godet, I.; Doctorman, S.; Wu, F.; Gilkes, D.M. Detection of Hypoxia in Cancer Models: Significance, Challenges, and Advances. Cells 2022, 11, 686. [Google Scholar] [CrossRef]
  118. Hubert, C.G.; Rivera, M.; Spangler, L.C.; Wu, Q.; Mack, S.C.; Prager, B.C.; Couce, M.; McLendon, R.E.; Sloan, A.E.; Rich, J.N. A Three-Dimensional Organoid Culture System Derived from Human Glioblastomas Recapitulates the Hypoxic Gradients and Cancer Stem Cell Heterogeneity of Tumors Found In Vivo. Cancer Res. 2016, 76, 2465–2477. [Google Scholar] [CrossRef]
  119. Lee, M.; Kim, Y.; Yu, S.J.; Lee, S.Y.; Son, J.G.; Lee, T.G.; Cho, Y.; Shin, J.H.; Lee, E.; Im, S.G. Surface Hydrophobicity Modulates the Key Characteristics of Cancer Spheroids through the Interaction with the Adsorbed Proteins. Adv. Funct. Mater. 2021, 31, 2100775. [Google Scholar] [CrossRef]
  120. Malhão, F.; Macedo, A.C.; Ramos, A.A.; Rocha, E. Morphometrical, Morphological, and Immunocytochemical Characterization of a Tool for Cytotoxicity Research: 3D Cultures of Breast Cell Lines Grown in Ultra-Low Attachment Plates. Toxics 2022, 10, 415. [Google Scholar] [CrossRef]
  121. Jubelin, C.; Muñoz-Garcia, J.; Griscom, L.; Cochonneau, D.; Ollivier, E.; Heymann, M.F.; Vallette, F.M.; Oliver, L.; Heymann, D. Three-Dimensional in Vitro Culture Models in Oncology Research. Cell Biosci. 2022, 12, 155. [Google Scholar] [CrossRef]
  122. Białkowska, K.; Komorowski, P.; Bryszewska, M.; Miłowska, K. Spheroids as a Type of Three-Dimensional Cell Cultures—Examples of Methods of Preparation and the Most Important Application. Int. J. Mol. Sci. 2020, 21, 6225. [Google Scholar] [CrossRef]
  123. Gai, T.; Zhang, Y.; Li, G.; Zhou, F.; He, C.; Wang, X.; Su, J. Engineered Hydrogel Microspheres for Spheroids and Organoids Construction. Chem. Eng. J. 2024, 498, 155131. [Google Scholar] [CrossRef]
  124. Lo Cicero, A.; Campora, S.; Lo Buglio, G.; Cinà, P.; Lo Pinto, M.; Scilabra, S.D.; Ghersi, G. Enhancing Therapeutic Efficacy through Degradation of Endogenous Extracellular Matrix in Primary Breast Tumor Spheroids. FEBS J. 2025, 292, 3494–3507. [Google Scholar] [CrossRef]
  125. Abbas, Z.N.; Al-Saffar, A.Z.; Jasim, S.M.; Sulaiman, G.M. Comparative Analysis between 2D and 3D Colorectal Cancer Culture Models for Insights into Cellular Morphological and Transcriptomic Variations. Sci. Rep. 2023, 13, 18380. [Google Scholar] [CrossRef] [PubMed]
  126. de Visser, K.E.; Joyce, J.A. The Evolving Tumor Microenvironment: From Cancer Initiation to Metastatic Outgrowth. Cancer Cell 2023, 41, 374–403. [Google Scholar] [CrossRef] [PubMed]
  127. Vakhshiteh, F.; Bagheri, Z.; Soleimani, M.; Ahvaraki, A.; Pournemat, P.; Alavi, S.E.; Madjd, Z. Heterotypic Tumor Spheroids: A Platform for Nanomedicine Evaluation. J. Nanobiotechnol. 2023, 21, 249. [Google Scholar] [CrossRef] [PubMed]
  128. Sheng, N.; Shindo, K.; Ohuchida, K.; Shinkawa, T.; Zhang, B.; Feng, H.; Yamamoto, T.; Moriyama, T.; Ikenaga, N.; Nakata, K.; et al. TAK1 Promotes an Immunosuppressive Tumor Microenvironment through Cancer-Associated Fibroblast Phenotypic Conversion in Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. 2024, 30, 5138–5153. [Google Scholar] [CrossRef]
  129. Cortini, M.; Macchi, F.; Reggiani, F.; Vitale, E.; Lipreri, M.V.; Perut, F.; Ciarrocchi, A.; Baldini, N.; Avnet, S. Endogenous Extracellular Matrix Regulates the Response of Osteosarcoma 3D Spheroids to Doxorubicin. Cancers 2023, 15, 1221. [Google Scholar] [CrossRef]
  130. Lazzari, G.; Nicolas, V.; Matsusaki, M.; Akashi, M.; Couvreur, P.; Mura, S. Multicellular Spheroid Based on a Triple Co-Culture: A Novel 3D Model to Mimic Pancreatic Tumor Complexity. Acta Biomater. 2018, 78, 296–307. [Google Scholar] [CrossRef]
  131. Zhao, Z.; Chen, X.; Dowbaj, A.M.; Sljukic, A.; Bratlie, K.; Lin, L.; Fong, E.L.S.; Balachander, G.M.; Chen, Z.; Soragni, A.; et al. Organoids. Nat. Rev. Methods Primers 2022, 2, 94. [Google Scholar] [CrossRef]
  132. Heydari, Z.; Moeinvaziri, F.; Agarwal, T.; Pooyan, P.; Shpichka, A.; Maiti, T.K.; Timashev, P.; Baharvand, H.; Vosough, M. Organoids: A Novel Modality in Disease Modeling. Bio-Des. Manuf. 2021, 4, 689–716. [Google Scholar] [CrossRef]
  133. Thorel, L.; Perréard, M.; Florent, R.; Divoux, J.; Coffy, S.; Vincent, A.; Gaggioli, C.; Guasch, G.; Gidrol, X.; Weiswald, L.B.; et al. Patient-Derived Tumor Organoids: A New Avenue for Preclinical Research and Precision Medicine in Oncology. Exp. Mol. Med. 2024, 56, 1531–1551. [Google Scholar] [CrossRef] [PubMed]
  134. Hillion, K.; Mahe, M.M. Redesigning Hydrogel Geometry for Enhanced Organoids. Nat. Methods 2022, 19, 1347–1348. [Google Scholar] [CrossRef] [PubMed]
  135. Kim, S.; Min, S.; Choi, Y.S.; Jo, S.H.; Jung, J.H.; Han, K.; Kim, J.; An, S.; Ji, Y.W.; Kim, Y.G.; et al. Tissue Extracellular Matrix Hydrogels as Alternatives to Matrigel for Culturing Gastrointestinal Organoids. Nat. Commun. 2022, 13, 1692. [Google Scholar] [CrossRef] [PubMed]
  136. Zhao, K.Y.; Du, Y.X.; Cao, H.M.; Su, L.Y.; Su, X.L.; Li, X. The Biological Macromolecules Constructed Matrigel for Cultured Organoids in Biomedical and Tissue Engineering. Colloids Surf. B Biointerfaces 2025, 247, 114435. [Google Scholar] [CrossRef]
  137. Aisenbrey, E.A.; Murphy, W.L. Synthetic Alternatives to Matrigel. Nat. Rev. Mater. 2020, 5, 539–551. [Google Scholar] [CrossRef]
  138. Tan, R.; Zhang, Z.; Ding, P.; Liu, Y.; Liu, H.; Lu, M.; Chen, Y.G. A Growth Factor-Reduced Culture System for Colorectal Cancer Organoids. Cancer Lett. 2024, 588, 216737. [Google Scholar] [CrossRef]
  139. Urbischek, M.; Rannikmae, H.; Foets, T.; Ravn, K.; Hyvönen, M.; de la Roche, M. Organoid Culture Media Formulated with Growth Factors of Defined Cellular Activity. Sci. Rep. 2019, 9, 6193. [Google Scholar] [CrossRef]
  140. Pinto, B.; Henriques, A.C.; Silva, P.M.A.; Bousbaa, H. Three-Dimensional Spheroids as In Vitro Preclinical Models for Cancer Research. Pharmaceutics 2020, 12, 1186. [Google Scholar] [CrossRef]
  141. Zhu, S.; Yin, J.; Lu, X.; Jiang, D.; Chen, R.; Cui, K.; He, W.; Huang, N.; Xu, G. Influence of Experimental Variables on Spheroid Attributes. Sci. Rep. 2025, 15, 9751. [Google Scholar] [CrossRef]
  142. Živković, Z.; Opačak-Bernardi, T. An Overview on Spheroid and Organoid Models in Applied Studies. Sci 2025, 7, 27. [Google Scholar] [CrossRef]
  143. Nwokoye, P.N.; Abilez, O.J. Bioengineering Methods for Vascularizing Organoids. Cell Rep. Methods 2024, 4, 100779. [Google Scholar] [CrossRef]
  144. Druzhkova, I.; Nikonova, E.; Ignatova, N.; Koryakina, I.; Zyuzin, M.; Mozherov, A.; Kozlov, D.; Krylov, D.; Kuznetsova, D.; Lisitsa, U.; et al. Effect of Collagen Matrix on Doxorubicin Distribution and Cancer Cells’ Response to Treatment in 3D Tumor Model. Cancers 2022, 14, 5487. [Google Scholar] [CrossRef]
  145. Aung, A.; Kumar, V.; Theprungsirikul, J.; Davey, S.K.; Varghese, S. An Engineered Tumor-on-a-Chip Device with Breast Cancer–Immune Cell Interactions for Assessing T-Cell Recruitment. Cancer Res. 2020, 80, 263–275. [Google Scholar] [CrossRef]
  146. Wan, L.; Neumann, C.A.; Leduc, P.R. Tumor-on-a-Chip for Integrating a 3D Tumor Microenvironment: Chemical and Mechanical Factors. Lab. Chip 2020, 20, 873–888. [Google Scholar] [CrossRef]
  147. Li, W.; Zhou, Z.; Zhou, X.; Khoo, B.L.; Gunawan, R.; Chin, Y.R.; Zhang, L.; Yi, C.; Guan, X.; Yang, M. 3D Biomimetic Models to Reconstitute Tumor Microenvironment In Vitro: Spheroids, Organoids, and Tumor-on-a-Chip. Adv. Healthc. Mater. 2023, 12, 2202609. [Google Scholar] [CrossRef]
  148. Park, S.E.; Georgescu, A.; Huh, D. Organoids-on-a-Chip. Science 2019, 364, 960–965. [Google Scholar] [CrossRef]
  149. Collins, T.; Pyne, E.; Christensen, M.; Iles, A.; Pamme, N.; Pires, I.M. Spheroid-on-Chip Microfluidic Technology for the Evaluation of the Impact of Continuous Flow on Metastatic Potential in Cancer Models in Vitro. Biomicrofluidics 2021, 15, 44103. [Google Scholar] [CrossRef]
  150. Uzabakiriho, P.C.; Jiajun, F.; Nguchu, B.A.; Iqbal, S.; Manishimwe, C.; Shaw, P. Spheroid-on-a-Chip Platforms for Tumor Microenvironment and Drug Development. Adv. Mater. Technol. 2025, 15, 2401821. [Google Scholar] [CrossRef]
  151. Goertzen, C.; Eymael, D.; Magalhaes, M. Three-Dimensional Quantification of Spheroid Degradation-Dependent Invasion and Invadopodia Formation. Biol. Proced. Online 2018, 20, 20. [Google Scholar] [CrossRef]
  152. Karve, K.; Poon, S.; Prinos, P.; Ailles, L. 3D Spheroid Invasion Assay for High-Throughput Screening of Small-Molecule Libraries. Methods Mol. Biol. 2023, 2706, 201–214. [Google Scholar] [CrossRef]
  153. Lamichhane, A.; Tavana, H. Three-Dimensional Tumor Models to Study Cancer Stemness-Mediated Drug Resistance. Cell. Mol. Bioeng. 2024, 17, 107–119. [Google Scholar] [CrossRef]
  154. Liao, J.; Qian, F.; Tchabo, N.; Mhawech-Fauceglia, P.; Beck, A.; Qian, Z.; Wang, X.; Huss, W.J.; Lele, S.B.; Morrison, C.D.; et al. Ovarian Cancer Spheroid Cells with Stem Cell-Like Properties Contribute to Tumor Generation, Metastasis and Chemotherapy Resistance through Hypoxia-Resistant Metabolism. PLoS ONE 2014, 9, e84941. [Google Scholar] [CrossRef]
  155. Ishtiah, A.A.; Yahaya, B.H. The Enrichment of Breast Cancer Stem Cells from MCF7 Breast Cancer Cell Line Using Spheroid Culture Technique. Methods Mol. Biol. 2022, 2429, 475–484. [Google Scholar] [CrossRef]
  156. Elberskirch, L.; Knoll, T.; Königsmark, R.; Renner, J.; Wilhelm, N.; von Briesen, H.; Wagner, S. Microfluidic 3D Intestine Tumor Spheroid Model for Efficient in Vitro Investigation of Nanoparticular Formulations. J. Drug Deliv. Sci. Technol. 2021, 63, 102496. [Google Scholar] [CrossRef]
  157. Petreus, T.; Cadogan, E.; Hughes, G.; Smith, A.; Pilla Reddy, V.; Lau, A.; O’Connor, M.J.; Critchlow, S.; Ashford, M.; Oplustil O’Connor, L. Tumour-on-Chip Microfluidic Platform for Assessment of Drug Pharmacokinetics and Treatment Response. Commun. Biol. 2021, 4, 1001. [Google Scholar] [CrossRef]
  158. Wang, Y.; Jeon, H. 3D Cell Cultures toward Quantitative High-Throughput Drug Screening. Trends Pharmacol. Sci. 2022, 43, 569–581. [Google Scholar] [CrossRef]
  159. Zanoni, M.; Pignatta, S.; Arienti, C.; Bonafè, M.; Tesei, A. Anticancer Drug Discovery Using Multicellular Tumor Spheroid Models. Expert. Opin. Drug Discov. 2019, 14, 289–301. [Google Scholar] [CrossRef]
  160. Nayak, P.; Bentivoglio, V.; Varani, M.; Signore, A. Three-Dimensional In Vitro Tumor Spheroid Models for Evaluation of Anticancer Therapy: Recent Updates. Cancers 2023, 15, 4846. [Google Scholar] [CrossRef]
  161. Saito, Y.; Muramatsu, T.; Kanai, Y.; Ojima, H.; Sukeda, A.; Hiraoka, N.; Arai, E.; Sugiyama, Y.; Matsuzaki, J.; Uchida, R.; et al. Establishment of Patient-Derived Organoids and Drug Screening for Biliary Tract Carcinoma. Cell Rep. 2019, 27, 1265–1276.e4. [Google Scholar] [CrossRef]
  162. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernández-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-Derived Organoids Model Treatment Response of Metastatic Gastrointestinal Cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef]
  163. Calpe, B.; Kovacs, W.J. High-Throughput Screening in Multicellular Spheroids for Target Discovery in the Tumor Microenvironment. Expert. Opin. Drug Discov. 2020, 15, 955–967. [Google Scholar] [CrossRef] [PubMed]
  164. Liang, J.; Zhao, D.K.; Yin, H.M.; Tian, T.Y.; Kang, J.K.; Shen, S.; Wang, J. Combinatorial Screening of Nanomedicines in Patient-Derived Cancer Organoids Facilitates Efficient Cancer Therapy. Nano Today 2025, 61, 102665. [Google Scholar] [CrossRef]
  165. Zuo, J.; Fang, Y.; Wang, R.; Liang, S. High-Throughput Solutions in Tumor Organoids: From Culture to Drug Screening. Stem Cells 2025, 43, sxae070. [Google Scholar] [CrossRef]
  166. Kim, M.; Yun, H.W.; Park, D.Y.; Choi, B.H.; Min, B.H. Three-Dimensional Spheroid Culture Increases Exosome Secretion from Mesenchymal Stem Cells. Tissue Eng. Regen. Med. 2018, 15, 427. [Google Scholar] [CrossRef]
  167. Haraszti, R.A.; Miller, R.; Stoppato, M.; Sere, Y.Y.; Coles, A.; Didiot, M.C.; Wollacott, R.; Sapp, E.; Dubuke, M.L.; Li, X.; et al. Exosomes Produced from 3D Cultures of MSCs by Tangential Flow Filtration Show Higher Yield and Improved Activity. Mol. Ther. 2018, 26, 2838–2847. [Google Scholar] [CrossRef]
  168. Bulati, M.; Gallo, A.; Zito, G.; Busà, R.; Iannolo, G.; Cuscino, N.; Castelbuono, S.; Carcione, C.; Centi, C.; Martucci, G.; et al. 3D Culture and Interferon-γ Priming Modulates Characteristics of Mesenchymal Stromal/Stem Cells by Modifying the Expression of Both Intracellular and Exosomal MicroRNAs. Biology 2023, 12, 1063. [Google Scholar] [CrossRef]
  169. Rovere, M.; Reverberi, D.; Arnaldi, P.; Palamà, M.E.F.; Gentili, C. Spheroid Size Influences Cellular Senescence and Angiogenic Potential of Mesenchymal Stromal Cell-Derived Soluble Factors and Extracellular Vesicles. Front. Bioeng. Biotechnol. 2023, 11, 1297644. [Google Scholar] [CrossRef]
  170. Xie, L.; Mao, M.; Zhou, L.; Zhang, L.; Jiang, B. Signal Factors Secreted by 2D and Spheroid Mesenchymal Stem Cells and by Cocultures of Mesenchymal Stem Cells Derived Microvesicles and Retinal Photoreceptor Neurons. Stem Cells Int. 2017, 2017, 2730472. [Google Scholar] [CrossRef]
  171. Kim, H.; Lee, M.J.; Bae, E.H.; Ryu, J.S.; Kaur, G.; Kim, H.J.; Kim, J.Y.; Barreda, H.; Jung, S.Y.; Choi, J.M.; et al. Comprehensive Molecular Profiles of Functionally Effective MSC-Derived Extracellular Vesicles in Immunomodulation. Mol. Ther. 2020, 28, 1628–1644. [Google Scholar] [CrossRef]
  172. Rocha, S.; Carvalho, J.; Oliveira, P.; Voglstaetter, M.; Schvartz, D.; Thomsen, A.R.; Walter, N.; Khanduri, R.; Sanchez, J.-C.; Keller, A.; et al. 3D Cellular Architecture Affects MicroRNA and Protein Cargo of Extracellular Vesicles. Adv. Sci. 2019, 6, 1800948. [Google Scholar] [CrossRef]
  173. Hu, S.; Li, Z.; Cores, J.; Huang, K.; Su, T.; Dinh, P.U.; Cheng, K. Needle-Free Injection of Exosomes Derived from Human Dermal Fibroblast Spheroids Ameliorates Skin Photoaging. ACS Nano 2019, 13, 11273–11282. [Google Scholar] [CrossRef] [PubMed]
  174. Hu, S.; Li, Z.; Lutz, H.; Huang, K.; Su, T.; Cores, J.; Dinh, P.U.C.; Cheng, K. Dermal Exosomes Containing MiR-218-5p Promote Hair Regeneration by Regulating β-Catenin Signaling. Sci. Adv. 2020, 6, eaba1685. [Google Scholar] [CrossRef] [PubMed]
  175. Dinh, P.U.C.; Paudel, D.; Brochu, H.; Popowski, K.D.; Gracieux, M.C.; Cores, J.; Huang, K.; Hensley, M.T.; Harrell, E.; Vandergriff, A.C.; et al. Inhalation of Lung Spheroid Cell Secretome and Exosomes Promotes Lung Repair in Pulmonary Fibrosis. Nat. Commun. 2020, 11, 1064. [Google Scholar] [CrossRef] [PubMed]
  176. Thippabhotla, S.; Zhong, C.; He, M. 3D Cell Culture Stimulates the Secretion of in Vivo like Extracellular Vesicles. Sci. Rep. 2019, 9, 13012. [Google Scholar] [CrossRef]
  177. Vera, N.; Acuña-Gallardo, S.; Grünenwald, F.; Caceres-Verschae, A.; Realini, O.; Acuña, R.; Lladser, A.; Illanes, S.E.; Varas-Godoy, M. Small Extracellular Vesicles Released from Ovarian Cancer Spheroids in Response to Cisplatin Promote the Pro-Tumorigenic Activity of Mesenchymal Stem Cells. Int. J. Mol. Sci. 2019, 20, 4972. [Google Scholar] [CrossRef]
  178. Giusti, I.; Poppa, G.; D’Ascenzo, S.; Esposito, L.; Vitale, A.R.; Calvisi, G.; Dolo, V. Cancer Three-Dimensional Spheroids Mimic In Vivo Tumor Features, Displaying “Inner” Extracellular Vesicles and Vasculogenic Mimicry. Int. J. Mol. Sci. 2022, 23, 11782. [Google Scholar] [CrossRef]
  179. Yang, J.; Zhang, Z.; Zhang, Y.; Ni, X.; Zhang, G.; Cui, X.; Liu, M.; Xu, C.; Zhang, Q.; Zhu, H.; et al. ZIP4 Promotes Muscle Wasting and Cachexia in Mice With Orthotopic Pancreatic Tumors by Stimulating RAB27B-Regulated Release of Extracellular Vesicles From Cancer Cells. Gastroenterology 2019, 156, 722–734.e6. [Google Scholar] [CrossRef]
  180. Jeon, T.J.; Kim, O.H.; Kang, H.; Lee, H.J. Preadipocytes Potentiate Melanoma Progression and M2 Macrophage Polarization in the Tumor Microenvironment. Biochem. Biophys. Res. Commun. 2024, 721, 150129. [Google Scholar] [CrossRef]
  181. Wang, J.; Wu, Y.; Guo, J.; Fei, X.; Yu, L.; Ma, S.; Wang, J.; Wu, Y.; Guo, J.; Fei, X.; et al. Adipocyte-Derived Exosomes Promote Lung Cancer Metastasis by Increasing MMP9 Activity via Transferring MMP3 to Lung Cancer Cells. Oncotarget 2017, 8, 81880–81891. [Google Scholar] [CrossRef]
  182. Donzelli, J.; Proestler, E.; Riedel, A.; Nevermann, S.; Hertel, B.; Guenther, A.; Gattenlöhner, S.; Savai, R.; Larsson, K.; Saul, M.J. Small Extracellular Vesicle-Derived MiR-574-5p Regulates PGE2-Biosynthesis via TLR7/8 in Lung Cancer. J. Extracell. Vesicles 2021, e12143. [Google Scholar] [CrossRef]
  183. Mullen, S.; Movia, D. The Role of Extracellular Vesicles in Non-Small-Cell Lung Cancer, the Unknowns, and How New Approach Methodologies Can Support New Knowledge Generation in the Field. Eur. J. Pharm. Sci. 2023, 188, 106516. [Google Scholar] [CrossRef]
  184. Relucenti, M.; Francescangeli, F.; De Angelis, M.L.; D’Andrea, V.; Miglietta, S.; Donfrancesco, O.; Li, X.; Chen, R.; Zeuner, A.; Familiari, G. A Different Exosome Secretion Pattern Characterizes Patient-Derived Colorectal Cancer Multicellular Spheroids and Their Mouse Xenografts. Biology 2022, 11, 1427. [Google Scholar] [CrossRef]
  185. Al Hrout, A.; Levesque, M.P.; Chahwan, R. Investigating the Tumor-Immune Microenvironment through Extracellular Vesicles from Frozen Patient Biopsies and 3D Cultures. Front. Immunol. 2023, 14, 1176175. [Google Scholar] [CrossRef] [PubMed]
  186. Schuster, M.; Braun, F.K.; Chiang, D.M.L.; Ludwig, C.; Meng, C.; Grätz, C.; Kirchner, B.; Proescholdt, M.; Hau, P.; Steinlein, O.K.; et al. Extracellular Vesicles Secreted by 3D Tumor Organoids Are Enriched for Immune Regulatory Signaling Biomolecules Compared to Conventional 2D Glioblastoma Cell Systems. Front. Immunol. 2024, 15, 1388769. [Google Scholar] [CrossRef] [PubMed]
  187. Santoro, J.; Carrese, B.; Peluso, M.S.; Coppola, L.; D’Aiuto, M.; Mossetti, G.; Salvatore, M.; Smaldone, G. Influence of Breast Cancer Extracellular Vesicles on Immune Cell Activation: A Pilot Study. Biology 2023, 12, 1531. [Google Scholar] [CrossRef] [PubMed]
  188. Dantzer, C.; Vaché, J.; Brunel, A.; Mahouche, I.; Raymond, A.A.; Dupuy, J.W.; Petrel, M.; Bioulac-Sage, P.; Perrais, D.; Dugot-Senant, N.; et al. Emerging Role of Oncogenic SS-Catenin in Exosome Biogenesis as a Driver of Immune Escape in Hepatocellular Carcinoma. eLife 2024, RP95191. [Google Scholar] [CrossRef]
  189. Eguchi, T.; Sogawa, C.; Okusha, Y.; Uchibe, K.; Iinuma, R.; Ono, K.; Nakano, K.; Murakami, J.; Itoh, M.; Arai, K.; et al. Organoids with Cancer Stem Cell-like Properties Secrete Exosomes and HSP90 in a 3D Nanoenvironment. PLoS ONE 2018, 13, e0191109. [Google Scholar] [CrossRef]
  190. Wang, Y.; Liu, J.; Ma, J.; Sun, T.; Zhou, Q.; Wang, W.; Wang, G.; Wu, P.; Wang, H.; Jiang, L.; et al. Exosomal CircRNAs: Biogenesis, Effect and Application in Human Diseases. Mol. Cancer 2019, 18, 116. [Google Scholar] [CrossRef]
  191. Liu, Y.; Ma, L.; Hua, F.; Min, Z.; Zhan, Y.; Zhang, W.; Yao, J. Exosomal CircCARM1 from Spheroids Reprograms Cell Metabolism by Regulating PFKFB2 in Breast Cancer. Oncogene 2022, 41, 2012–2025. [Google Scholar] [CrossRef]
  192. Xu, F.; Wang, K.; Zhu, C.; Fan, L.; Zhu, Y.; Wang, J.F.; Li, X.; Liu, Y.; Zhao, Y.; Zhu, C.; et al. Tumor-Derived Extracellular Vesicles as a Biomarker for Breast Cancer Diagnosis and Metastasis Monitoring. iScience 2024, 27, 109506. [Google Scholar] [CrossRef]
  193. Mathew, M.; Zade, M.; Mezghani, N.; Patel, R.; Wang, Y.; Momen-Heravi, F. Extracellular Vesicles as Biomarkers in Cancer Immunotherapy. Cancers 2020, 12, 2825. [Google Scholar] [CrossRef] [PubMed]
  194. Huang, L.; Bockorny, B.; Paul, I.; Akshinthala, D.; Frappart, P.O.; Gandarilla, O.; Bose, A.; Sanchez-Gonzalez, V.; Rouse, E.E.; Lehoux, S.D.; et al. PDX-Derived Organoids Model in Vivo Drug Response and Secrete Biomarkers. JCI Insight 2020, 5, e135544. [Google Scholar] [CrossRef] [PubMed]
  195. Bari, E.; Perteghella, S.; Catenacci, L.; Sorlini, M.; Croce, S.; Mantelli, M.; Avanzini, M.A.; Sorrenti, M.; Torre, M.L. Freeze-Dried and GMP-Compliant Pharmaceuticals Containing Exosomes for Acellular Mesenchymal Stromal Cell Immunomodulant Therapy. Nanomedicine 2019, 14, 753–765. [Google Scholar] [CrossRef] [PubMed]
  196. Son, J.P.; Kim, E.H.; Shin, E.K.; Kim, D.H.; Sung, J.H.; Oh, M.J.; Cha, J.M.; Chopp, M.; Bang, O.Y. Mesenchymal Stem Cell-Extracellular Vesicle Therapy for Stroke: Scalable Production and Imaging Biomarker Studies. Stem Cells Transl. Med. 2023, 12, 459–473. [Google Scholar] [CrossRef]
  197. Zeöld, A.; Sándor, G.O.; Kiss, A.; Soós, A.Á.; Tölgyes, T.; Bursics, A.; Szűcs, Á.; Harsányi, L.; Kittel, Á.; Gézsi, A.; et al. Shared Extracellular Vesicle MiRNA Profiles of Matched Ductal Pancreatic Adenocarcinoma Organoids and Blood Plasma Samples Show the Power of Organoid Technology. Cell. Mol. Life Sci. 2021, 78, 3005–3020. [Google Scholar] [CrossRef]
  198. Xiao, W.; Pahlavanneshan, M.; Eun, C.Y.; Zhang, X.; DeKalb, C.; Mahgoub, B.; Knaneh-Monem, H.; Shah, S.; Sohrabi, A.; Seidlits, S.K.; et al. Matrix Stiffness Mediates Pancreatic Cancer Chemoresistance through Induction of Exosome Hypersecretion in a Cancer Associated Fibroblasts-Tumor Organoid Biomimetic Model. Matrix Biol. Plus 2022, 14, 100111. [Google Scholar] [CrossRef]
  199. Xu, K.; Shen, R.; Zhang, L.; Gao, X.; Wang, X.; Zhang, C.; Chen, X.; Wang, X. Pancreatic Cancer-Derived Extracellular Vesicles Enriched with MiR-223-5p Promote Skeletal Muscle Wasting Associated with Cachexia. Adv. Sci. 2025. [Google Scholar] [CrossRef]
  200. Buenafe, A.C.; Dorrell, C.; Reddy, A.P.; Klimek, J.; Marks, D.L. Proteomic Analysis Distinguishes Extracellular Vesicles Produced by Cancerous versus Healthy Pancreatic Organoids. Sci. Rep. 2022, 12, 3556. [Google Scholar] [CrossRef]
  201. Nagai, H.; Kuroha, M.; Handa, T.; Karasawa, H.; Ohnuma, S.; Naito, T.; Moroi, R.; Kanazawa, Y.; Shiga, H.; Hamada, S.; et al. Comprehensive Analysis of MicroRNA Profiles in Organoids Derived from Human Colorectal Adenoma and Cancer. Digestion 2021, 102, 860–869. [Google Scholar] [CrossRef]
  202. Szvicsek, Z.; Oszvald, Á.; Szabó, L.; Sándor, G.O.; Kelemen, A.; Soós, A.Á.; Pálóczi, K.; Harsányi, L.; Tölgyes, T.; Dede, K.; et al. Extracellular Vesicle Release from Intestinal Organoids Is Modulated by Apc Mutation and Other Colorectal Cancer Progression Factors. Cell. Mol. Life Sci. 2019, 76, 2463–2476. [Google Scholar] [CrossRef] [PubMed]
  203. Taha, E.A.; Sogawa, C.; Okusha, Y.; Kawai, H.; Oo, M.W.; Elseoudi, A.; Lu, Y.; Nagatsuka, H.; Kubota, S.; Satoh, A.; et al. Knockout of MMP3 Weakens Solid Tumor Organoids and Cancer Extracellular Vesicles. Cancers 2020, 12, 1260. [Google Scholar] [CrossRef]
  204. Sasaki, A.; Kuroha, M.; Tosa, M.; Takahashi, S.; Oomori, S.; Nomura, E.; Kikuchi, T.; Onodera, M.; Sato, Y.; Miyazawa, T.; et al. Evaluation of Organoid-Derived Exosomal MicroRNA as Liquid Biopsy for Colorectal Cancer: A Multicenter Cross-Sectional Study. Clin. Transl. Sci. 2025, 18, e70270. [Google Scholar] [CrossRef]
  205. Tauro, B.J.; Greening, D.W.; Mathias, R.A.; Mathivanan, S.; Ji, H.; Simpson, R.J. Two Distinct Populations of Exosomes Are Released from LIM1863 Colon Carcinoma Cell-Derived Organoids. Mol. Cell. Proteom. 2013, 12, 587–598. [Google Scholar] [CrossRef]
  206. Sándor, G.O.; Soós, A.Á.; Lörincz, P.; Rojkó, L.; Harkó, T.; Bogyó, L.; Tölgyes, T.; Bursics, A.; Buzás, E.I.; Moldvay, J.; et al. Wnt Activity and Cell Proliferation Are Coupled to Extracellular Vesicle Release in Multiple Organoid Models. Front. Cell Dev. Biol. 2021, 9, 670825. [Google Scholar] [CrossRef]
Figure 1. Extracellular vesicle biogenesis. Two main types of EVs are distinguished based on their biogenesis. Exosomes (30–150 nm) are generated through the endocytic pathway and are released via exocytosis of the multivesicular bodies (MVBs). Late endosomes either fuse with lysosomes for degradation or fuse with the plasma membrane to release the intraluminal vesicles that are called exosomes. Microvesicles (100–1000 nm) are released through budding from the plasma membrane. EVs can be internalized by recipient cells through endocytosis, receptor-mediated signaling pathways, or membrane fusion.
Figure 1. Extracellular vesicle biogenesis. Two main types of EVs are distinguished based on their biogenesis. Exosomes (30–150 nm) are generated through the endocytic pathway and are released via exocytosis of the multivesicular bodies (MVBs). Late endosomes either fuse with lysosomes for degradation or fuse with the plasma membrane to release the intraluminal vesicles that are called exosomes. Microvesicles (100–1000 nm) are released through budding from the plasma membrane. EVs can be internalized by recipient cells through endocytosis, receptor-mediated signaling pathways, or membrane fusion.
Ijms 26 07104 g001
Figure 2. Comprehensive role of EVs in cancer. Tumor-derived EVs contribute to multiple cancer-related processes, including tumor growth, pre-metastatic niche (PMN) formation, immune evasion, angiogenesis, metastasis, drug resistance, and drug delivery.
Figure 2. Comprehensive role of EVs in cancer. Tumor-derived EVs contribute to multiple cancer-related processes, including tumor growth, pre-metastatic niche (PMN) formation, immune evasion, angiogenesis, metastasis, drug resistance, and drug delivery.
Ijms 26 07104 g002
Figure 3. Representative models of tumor spheroids, multicellular tumor spheroids (MCTS), tumor organoids, and intestinal tumor organoids. Tumor spheroids are three-dimensional aggregates of cancer cells, while multicellular tumor spheroids incorporate additional cell types, such as cancer-associated fibroblasts (CAFs), endothelial cells, immune cells, or adipocytes, to better replicate the tumor microenvironment and intercellular interactions. Tumor organoids are more complex structures with well-defined cell organization, vascularization, and an endogenous or additional complex extracellular matrix (ECM). Both models exhibit a characteristic oxygen, pH, and nutrient gradient from the outer proliferative layer to the inner core. The central regions often become hypoxic, closely resembling conditions within solid tumors. Some tumor organoids, like intestinal ones, present a specific polarization or cells with an inner lumen and mesenchymal cells in the external part and do not present the different layers.
Figure 3. Representative models of tumor spheroids, multicellular tumor spheroids (MCTS), tumor organoids, and intestinal tumor organoids. Tumor spheroids are three-dimensional aggregates of cancer cells, while multicellular tumor spheroids incorporate additional cell types, such as cancer-associated fibroblasts (CAFs), endothelial cells, immune cells, or adipocytes, to better replicate the tumor microenvironment and intercellular interactions. Tumor organoids are more complex structures with well-defined cell organization, vascularization, and an endogenous or additional complex extracellular matrix (ECM). Both models exhibit a characteristic oxygen, pH, and nutrient gradient from the outer proliferative layer to the inner core. The central regions often become hypoxic, closely resembling conditions within solid tumors. Some tumor organoids, like intestinal ones, present a specific polarization or cells with an inner lumen and mesenchymal cells in the external part and do not present the different layers.
Ijms 26 07104 g003
Figure 4. Comparison of 2D and 3D cell culture systems: morphological features and extracellular vesicle (EV) differences. Key distinctions between 2D and 3D culture models in terms of cell morphology, nutrient and oxygen gradients, and the extent of cell–cell and cell–extracellular matrix (ECM) interactions. It also highlights the relative advantages of each system for EV production.
Figure 4. Comparison of 2D and 3D cell culture systems: morphological features and extracellular vesicle (EV) differences. Key distinctions between 2D and 3D culture models in terms of cell morphology, nutrient and oxygen gradients, and the extent of cell–cell and cell–extracellular matrix (ECM) interactions. It also highlights the relative advantages of each system for EV production.
Ijms 26 07104 g004
Table 1. EVs from tumor 3D spheroids and organoids.
Table 1. EVs from tumor 3D spheroids and organoids.
TumorCancer Cells3D Model3D EV Isolation MethodFindingReferences
Gastric cancerMKN74 MKN45SpheroidDifferential ultracentrifugationUpregulation of microRNAs and downregulation of proteins in 3D EVs[172]
Cervical cancerHelaSpheroidFiltrationEV small RNAs[176]
Ovarian cancerHeyA8
Ovcar3
SpheroidDifferential ultracentrifugationPro-angiogenetic role[177]
Ovarian cancerCABA ISpheroidDifferential ultracentrifugation“Inner” EVs[178]
Pancreatic cancerAsPC-1
BxPC-3
SpheroidIsolation kitZIP4 knockdown reduced EVs’ pancreatic cancer release[179]
MelanomaB16SpheroidN/AInfluence of preadipocytes in melanoma growth[180]
Lung cancer3LL
A549
SpheroidDifferential ultracentrifugationModulation of tumor microenvironment[181]
Lung cancerA549
H1650
2106T
SpheroidDifferential ultracentrifugationRole of miR-574-5p in prostaglandin H2 regulation[182]
Colorectal cancerPrimarySpheroidN/AMultilayer spheroids release more EVs[184]
GlioblastomaIDH wild-type (CNS WHO grade 4)-derived models (BTIC10, -13, -131, -18, -129, -155)OrganoidPrecipitation and immunoaffinityComparison between EVs released from 2D and 3D models[186]
Breast cancerHS578T
BT474
SpheroidDifferential ultracentrifugationEffects of EVs breast cancer on PBMC from healthy donors[187]
Hepatocellular carcinomaHepG2
Huh7
SpheroidDifferential ultracentrifugationβ-catenin decreases EV release and immune cell infiltration[188]
Prostatic adenocarcinomaPC-3SpheroidDifferential ultracentrifugation
Filtration
Secretion of HSP90 and EpCAM[189]
Breast cancerMDA-MB-231SpheroidDifferential ultracentrifugationcircCARM1 promotes breast cancer proliferation and glycolysis[191]
Pancreatic ductal adenocarcinomaPrimary cellsSpheroidFiltration and ultracentrifugationNew biomarkers[194]
Pancreatic ductal adenocarcinomaPDAC cell lines (derived from primary tumors)OrganoidDifferential ultracentrifugationmiRNA EVs released with matched patient plasma and extracellular matrix remodeling[197]
Pancreatic ductal adenocarcinomaMouse-derived organoidsOrganoidIsolation kit3D biomimetic PDAC model with integrated CAF[198]
Pancreatic ductal adenocarcinomaPatient-derived organoidsOrganoidDifferential ultracentrifugationAbsorption of miRNA in PDAC-derived EVs by skeletal muscles and the role in cachexia[199]
Pancreatic ductal adenocarcinomaPatient-derived organoidsOrganoidIsolation kitDifferences between EVs from PDAC organoids and healthy pancreatic organoids[200]
Colorectal cancerPatient-derived organoidsOrganoidDifferential ultracentrifugationThe role of miR-1246 in promoting proliferation[201]
Colorectal cancerMouse- and patient-derived organoidsOrganoidIsolation kitAPC mutation and collagen deposition enhance EV release[202]
Colorectal cancerLuM1 cell lineOrganoidDifferential centrifugation and concentrationMMP3 knockout led to the additional release of EVs from organoids[203]
Colorectal cancerPatient-derived organoidsOrganoidDifferential ultracentrifugation and filtrationmiR-4284, miR-5100, miR-1246, miR-1290 elevated[204]
Colorectal cancerHuman colon carcinoma LIM1863 cellsOrganoidIsolation kitEVs isolated from apical and basolateral region have distinct proteomic profiles[205]
Pancreatic ductal adenocarcinoma
Lung bronchiolar
Lung adenocarcinoma
Human PDAC organoids
Mouse pancreas ductal and lung organoids
Human bronchiolar and LUAD organoids
OrganoidDifferential ultracentrifugationWnt signaling is tightly coupled to cell proliferation and EV secretion in lung adenocarcinoma but disrupted in PDAC[206]
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.

Share and Cite

MDPI and ACS Style

Campora, S.; Lo Cicero, A. The 3D Language of Cancer: Communication via Extracellular Vesicles from Tumor Spheroids and Organoids. Int. J. Mol. Sci. 2025, 26, 7104. https://doi.org/10.3390/ijms26157104

AMA Style

Campora S, Lo Cicero A. The 3D Language of Cancer: Communication via Extracellular Vesicles from Tumor Spheroids and Organoids. International Journal of Molecular Sciences. 2025; 26(15):7104. https://doi.org/10.3390/ijms26157104

Chicago/Turabian Style

Campora, Simona, and Alessandra Lo Cicero. 2025. "The 3D Language of Cancer: Communication via Extracellular Vesicles from Tumor Spheroids and Organoids" International Journal of Molecular Sciences 26, no. 15: 7104. https://doi.org/10.3390/ijms26157104

APA Style

Campora, S., & Lo Cicero, A. (2025). The 3D Language of Cancer: Communication via Extracellular Vesicles from Tumor Spheroids and Organoids. International Journal of Molecular Sciences, 26(15), 7104. https://doi.org/10.3390/ijms26157104

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop