REVIEW article

Front. Genet., 01 May 2025

Sec. Cancer Genetics and Oncogenomics

Volume 16 - 2025 | https://doi.org/10.3389/fgene.2025.1545017

This article is part of the Research Topic3D in Vitro Models - A New Era in Cancer ResearchView all articles

In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation

  • 1Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
  • 2Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
  • 3KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
  • 4Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden

Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.

1 Introduction

Colorectal cancer (CRC), which includes colon and/or rectum cancer, is a major public health concern because it is the third most diagnosed and second most lethal cancer worldwide (Morgan et al., 2023). CRC accounted for approximately 9% of all cancer-related deaths in 2022 (Bray et al., 2024), and the incidence of this disease is expected to rise to 2.5 million new cases by 2035 (Dekker et al., 2019). CRC is related to environmental factors (e.g., smoking, diet, obesity, alcohol), immune system dysregulation, or genetics, such as mutations of microsatellite instability (MSI) genes MSH1, MLH1, and MSH6, adenomatous polyposis coli (APC), or nucleotide-binding oligomerization domain 2 (NOD2) (Lawes et al., 2005). CRC begins with the transformation of a normal colonic crypt into a hyperproliferation and then into a benign adenomatous polyp (Figure 1). A small proportion of these adenomatous polyps will progress to advanced adenocarcinoma, causing cancerous growth (malignant neoplasia) and metastases. Progressive transition, also known as ‘multistep carcinogenesis’, occurs during CRC development, and unique genetic modifications of tumor suppressors or oncogenes correspond to each stage (Fearon, 2011). Mutations with the tumor suppressor genes like APC and TP53 are usually associated with CRC (Alzahrani et al., 2021). Oncogenes involved in CRC include RAS genes (KRAS, HRAS, and NRAS), BRAF, AKT1, EGFR, PIK3CA, MYC, and JAK (Menyhart et al., 2019).

Figure 1
www.frontiersin.org

Figure 1. CRC development and transformation in different stages from adenomatous polyps to metastases. Four stages in the development of CRC carcinogenesis occur: initiation, promotion, progression, and metastasis. The most common metastatic site is the liver, followed by the lung and bone. Created with BioRender.com.

The overall 5-year survival rate for CRC patients is 65%, although the individual prognosis is largely determined by whether the patient develops metastases during the disease. Within 5 years, up to 60% of all CRC patients develop metastatic CRC (mCRC), resulting in a dismal survival rate of less than 15% (Siegel et al., 2023). Approximately 15% of CRC patients have liver metastases at the time of diagnosis (Manfredi et al., 2006), and an additional 16%–20% develop hepatic metastases within the first 3 years (Kemeny, 2007). Lung metastases are less common and mostly occur in patients with rectal cancer (Mitry et al., 2010). These data highlight the importance of more effective treatments for both CRC and mCRC.

Clinically, the CRC staging system defines tumor severity on the basis of histopathological features and helps estimate the probability of disease relapse in patients with locoregional CRC (stages I–III) and those diagnosed with metastasis (stage IV) (Cañellas-Socias et al., 2024). Owing to limits in early detection, a considerable majority of patients are in an advanced stage when they are diagnosed. The most common treatment strategy for early-stage CRC is surgery followed by chemotherapy or, in certain situations, radiotherapy in the rectum. CRC can also be divided into four groups based on its consensus molecular subtypes (Figure 2) (Guinney et al., 2015). The first group contains disrupted MSI and activated immune components. The second involves epithelial differentiation and MYC signaling activation. The third group shows metabolic dysregulation, while the fourth group shows the expression of genes related to stromal invasion, epithelial–mesenchymal transition (EMT), transforming growth factor-beta (TGF-β), and angiogenesis.

Figure 2
www.frontiersin.org

Figure 2. CRC are divided into four different groups based on the consensus molecular subtypes. The first group is characterized by MSI, high mutations of BRAF and activated immune components. The second involves epithelial differentiation and MYC mutations, causing high activity of these intracellular signaling pathways. It also features increased expression of EGFR, TP53 and APC. The third group shows metabolic dysregulation with increased activity in glutaminolysis and lipidogenesis, enriched with KRAS activating mutations. The fourth group show the expression of genes related to stromal invasion, epithelial–mesenchymal transition (EMT), transforming growth factor-beta (TGF-β), and angiogenesis. Created with BioRender.com.

The introduction of 3D culture systems to in vitro modelling is expected to have a revolutionary impact on CRC research. With 3D culture options, it is possible to better replicate the human tumor and the complex tumor microenvironment (TME) of CRC, which is crucial in CRC development, progression, and metastasis. By mimicking the in vivo conditions of the tumor more closely by including microenvironment components such as stroma, immune cells and various extracellular matrix (ECM) glycoproteins, i.e., collagen, fibronectin, and laminins, 3D models offer significant advantages for the understanding of tumor biology, drug responses, and potential therapeutic strategies. A deep comprehension of the TME components is expected to allow development of effective CRC treatments. Recently, several 3D in vitro models have been developed specifically for CRC, including but not limited to spheroids, patient-derived organoids (PDO), bioprinted models, microfluidic models and scaffold-based models (Vitale et al., 2024; Li J. et al., 2024; Mo et al., 2022). 3D models became the most preferred approach in cancer research to fill the gap between “absolute in vitro” and “true in vivo” (Abbas et al., 2023). This review addresses the challenges of mimicking the TME components in an in vitro 3D model for CRC. It also underscores the necessity of developing more physiologically relevant 3D models. The importance of the role of the different ECM components in 3D models of CRC are discussed with an emphasis on how the TME affects tumor growth and treatment. Support is provided that there is a great need for in vitro 3D models that mimic the interactions within the TME, essential for screening for novel anticancer therapies. Furthermore, the knowledge of the TME and how nanotechnology can be integrated is highlighted. Therefore, a deep comprehension of the TME components is explored to find the better way for an effective CRC treatment. This review aims to bridge the gap between current model systems and the intricate biology of CRC.

2 Current in vitro models of CRC and mCRC

2D cell culture is one of the most common methods for studying CRC cells in vitro. However, these cell cultures involve the growth of a standardized monolayer, and some of the functionally important epithelial cell characteristics are not developed, potentially yielding results with little physiological value. For example, culturing several CRC epithelial cell types on an ultra-stiff plastic matrix induces cell proliferation and mesenchymal-like malignant phenotypes (Chaudhuri et al., 2014). Additionally, the lack of signals given by other stroma cell types may modify the morphological organization or response of intestinal cells (Paduch et al., 2010), or make sensitive tumor cell lines resistant to targeted drugs (Straussman et al., 2012). As a result, several analyses on cellular processes, such as proliferation, differentiation or apoptosis may be aberrant or not entirely true (Edmondson et al., 2014).

Spheroids are multicellular self-assembled 3D structures that can be generated via either scaffold-free or scaffold-based culture methods (Ramos et al., 2023). Scaffold-free spheroid formation methods include the hanging drop method, and culture using non-adherent surfaces (Reidy et al., 2021; Hasterok et al., 2023). Scaffold-free 3D cell cultures rely on the fact that cells adhere to each other when no other options are available. The hanging drop method uses surface tension and gravitational force to form spheroids by gathering cells in the lower part of a droplet hanging from an inverted tray (Jensen and Teng, 2020). Due to the ability of many adherent tumor cells to aggregate into higher cell densities, 3D spheroids have been widely used as cancer models in vitro.

The scaffold-based 3D culture methods typically use some type of hydrogel, and cell cultures can be achieved with either natural hydrogels (e.g., chitosan, alginate, silk fibroin) or synthetic hydrogels (e.g., polyethylene glycol (PEG), polylactic acid, and polyglycolic acid) (Ramos et al., 2023). Hydrogels are cross-linked networks formed of hydrophilic polymers attached through physical, electrostatic, or covalent interactions. Most hydrogels of synthetic polymers exhibit versatile biophysical, mechanical, and biological properties (Habanjar et al., 2021). However, synthetic polymers are unable to provide the biochemical signals necessary to “communicate” with the cell. To overcome this limitation, synthetic polymers can be functionalized by adding signalling biomolecules, such as peptides, growth factors, and glycans. Various natural polymers, of either animal or plant origin, can be used to form hydrogels. Examples of such natural polymers are animal-derived collagen, laminins, and fibrinogen, microbial-derived hyaluronic acid (HA) or plant-derived alginate and gellan gum (Bray et al., 2015). Collagen-based hydrogels are often used for 3D culture, since collagen is a major constituent of both the basement membrane and interstitial ECM. The commercially available Matrigel ®(Corning, Corning, NY, United States of America), is derived from mice with induced Engelberth–Holm–Swarm chondrosarcoma rich in ECM components. Those include laminin-111, type IV collagen, entactins and perlecan, as well as soluble growth factors, such as fibroblast growth factor (FGF), epidermal growth factor (EGF), transformative growth factor beta (TGF-beta), and MMPs (Marchini and Gelain, 2022). Since animal-derived ECMs substrates, such as collagen and Matrigel ®, are characterized by a poorly defined composition and batch-to-batch variability, increasing efforts have been made to develop bioinspired or fully-synthetic materials that could replace naturally-derived matrices, aiming for protocols that are more reproducible and translatable into clinical applications. For example, recombinant versions of ECM proteins such as laminins are commercially avalaible (BioLamina). Due to the more complicated post-translational modifications needed to obtain structurally functional collagen, the recombinant versions of fibrillar collagens are still difficult to obtain from recombinant processes (Fertala, 2020). Typical cell sources for CRC in vitro models are various immortalized cancer cell lines or dissociated cell clusters from resected tumors. There are a huge number of available primary CRC cell lines (Berg et al., 2017) whereof HCT116, HT29 and SW620 are commonly used for representation of the primary tumor. In contrast, established mCRC cell lines from the liver are less common, but exist (Boot et al., 2016).

In recent years, more and more investigations have been performed using spheroids formed from fresh primary cancer cells (Jeppesen et al., 2017) and patient-derived explants (Vitale et al., 2024). However, the typically limited life span (∼72 h) of primary cultures hinders longer studies of e.g., drug treatment effects. Lately, 3D culture into so called organoids have been more and more developed. Organoids refer to more complex cellular arrangements of organ-specific cells, that can be derived from pluripotent stem cells (PSCs), adult stem cells, or somatic cells of human tissues such as cancer cells (Ahmad Zawawi et al., 2024). Organoids mimic complex key structure, function, and biology of organs or tissues from which they are derived. The cells exhibit extended proliferation and differentiation capabilities over a long-term of culture, depending on specific culture medium used, and possess certain characteristics reminiscent of the organ or tumor from where they derived from. Pioneering development for organoid establishment was done by embedding isolated intestinal crypts or Lgr5-expressing intestinal stem cells into ECM and adding a special culture medium that contains essential growth factors, to obtain intestinal organoids (Sato et al., 2009). Since CRC, especially differentiated types that form ductal structures, are composed of a heterogeneous population of undifferentiated cancer stem cells (CSCs) and differentiated cells, constituting a hierarchical structure, a lot of efforts have been made to understand the role of CSCs in organoids from intestinal cells (Kawai et al., 2020). By the use of similar culture protocols, PDOs can be obtained from individual patients with high success rate, short culture period and unlimited expansion, which can highly recapitulate physiology of the original tumor (Guinney et al., 2015). Studies have shown that PDOs can not only be used to explore tumor biological characteristics in basic research,but also be used as a preclinical model to predict patients’ response to treatment (Mo et al., 2022). Indeed, CRC organoids retain tumor gene characteristics, heterogeneity, and intratumoral cellular heterogeneity, almost perfectly replicating CRC histological features, as compared with cancer cell lines and animal models (Li J. et al., 2024). However, 3D cultures derived from CRC patients consist of phenotypically heterogeneous and interchangeable spheroid-forming cells and can therefore show different growth rates and drug sensitivity (Coppo et al., 2023).

Accumulating evidence suggests that CRC cells represent phenotypically dynamic (rather than static), heterogeneous cell populations that display cell plasticity characteristics. Moreover, organoids derived from CRC and metastatic tissues also showed preserved genetic diversity and morphological stability (Buzzelli et al., 2018). Tumor organoids have the ability to more accurately simulate the complex TME and can be utilized for screening anti-tumor drugs, predicting patient responses to drug therapy, and guiding personalized treatment plans (Qu et al., 2024).

The main advantages and limits of different pre-clinical in vitro models used in CRC are summarized in Table 1 (Reidy et al., 2021; Habanjar et al., 2021; Qu et al., 2024; Liu Y. et al., 2024). The strengths of 3D in vitro CRC models are extensively reported in this review, but weaknesses also exist. The primary challenges are the technical complexity and cost since the creation and maintenance of 3D cultures is more complex and expensive than traditional 2D models, limiting their utility in some settings. The scalability issues are also another problem because some 3D models may not easily scale for large-scale drug screening or clinical applications. Additionally, the limited standardization of protocols for in vitro 3D models leads to variability in results across different laboratory and research projects. Furthermore, despite the advancements, many 3D models fail to fully replicate the intricate TME since they are missing key components like blood vessels, stroma, immune cells, and the ECM.

Table 1
www.frontiersin.org

Table 1. Comparison between the different pre-clinical models.

3 The natural tumor microenvironment

Cancer research has expanded its focus to incorporate the importance of the TME, which is a dynamic ecosystem encompassing both cellular and acellular components (Palucka and Coussens Lisa, 2016). The cellular components of the TME are a heterogeneous population of cancerous and non-cancerous cells. Non-cancerous cells are mainly immune cells and stromal cells. The TME acellular compartment includes the ECM, growth factors, cytokines, and other soluble mediators (Walker et al., 2018). This complex network of interactions is critical for cancer progression and the response to therapy. The interaction of cancer cells with their microenvironment is essential not only for the primary CRC tumor but also for the metastatic scenario. The invasion of cancer cells from the primary tumor via the ECM of the stroma is a critical stage that is driven by a multitude of biochemical and biophysical cues in the acellular ECM (Peela et al., 2017). Upon entry of the circulating CRC cells from the blood to the liver, several basic steps are implicated in the formation of liver metastases. Hepatic stellate cells (HSCs) that are activated by the pro-inflammatory cascade play vital roles in liver physiology and fibrogenesis and produce ECM-related proteins (Tsilimigras et al., 2023; Chandra et al., 2021). Neo-vascularization is mediated by vascular endothelial growth factor (VEGF) produced by the tumor cells or activated liver-resident macrophage-like Kupffer cells (Figure 3). Oxygen and nutrients are thereby supplied to the tumor cells, and the metastatic cells can multiply and enlarge to form detectable tumors. Moreover, in hepatic mCRC, increased fructose metabolism was observed through the upregulation of the enzyme aldolase B (ALDOB), providing additional fuel for metastatic growth (Bu et al., 2018). Thus, as the metabolic hub of the entire organism, the liver appears to create a unique environment that allows or forces cancer cells to engage in certain metabolic activities for their colonization. In addition, oncogenic KRAS and transforming growth factor-beta (TGF-β) have been shown to promote and maintain CRC metastasis via the regulation of immunity and cell–cell contact (Boutin et al., 2017). Understanding the molecular mechanisms that enable metastasis, determining and validating the key targets that maintain metastasis, and identifying how these targets might also influence primary tumor maintenance are all critical knowledge gaps, that could be filled by the use of advanced in vitro 3D models.

Figure 3
www.frontiersin.org

Figure 3. 3D models mimicking the TME of CRC and mCRC compartments. A complex network of cellular and ECM microenvironmental interactions is critical for cancer progression, and is essential not only for the primary CRC tumor, but also for the metastatic scenario. Except for the immune and stromal cells, liver sinusoidal endothelial cells, Kupffer cells and other liver macrophages are necessary for maintaining the mCRC tissue environment in the liver. Created with BioRender.com.

3.1 The extracellular matrix in the tumor microenvironment

The ECM compartment regulates cancer cell behaviour such as proliferation, adhesion, migration, and differentiation (Bonnans et al., 2014) and its function in cancer and metastasis has been extensively discussed (Kai et al., 2019). The ECM constitutes over 300 components, the majority of which are proteins and glycoproteins (incl. collagens, laminins, elastin, fibronectin, and tenascins), proteoglycans (incl. heparan sulphate), and glycosaminoglycans (incl. HA). At the structural level, there are two distinct ECM compartments to consider: the interstitial compartment, which is a network mostly composed of fibrillar collagens and non-collagenous glycoproteins, and the basement membrane (BM), which separates the epithelial layer from the underlying tissue (Manou et al., 2019) (Nyström, 2021). The BM is made up of a fibril mesh mostly composed of type IV collagen and laminins. Laminins are composed of three polypeptides (α1-5, β1-4 and γ1-3) in diverse combinations, resulting in fifteen known variations forming a cross-like structure (Aumailley, 2013).

As tumor progression occurs, the cancer cells subsequently interact with a completely different niche, the tumor-associated stroma, which is composed of activated mesenchymal cells and a collagen I-rich ECM with a peculiar topography. Research has demonstrated that the ECM of CRC TME dynamically shifts during each stage of development, indicating its importance in cancer progression (Li ZL. et al., 2020). In addition to changes in the composition of the ECM, the stiffness of the ECM increases as CRC advances (Kawano et al., 2015), and increased stiffness may limit drug delivery and enhance resistance (Schrader et al., 2011). Studies on CRC have shown that changes in the ECM can either limit tumor development or promote tumor progression (Miura et al., 1993). These mechanical qualities emerge as fundamental drivers of cellular activity since, for example, increasing matrix stiffness can lead to an increase in focal adhesions, rupture of adherent junctions and accelerated proliferation (Paszek et al., 2005). Therefore, an in vitro CRC model including ECM, either incorporated into their architecture or provided by cells that release ECM components, would be beneficial.

Collagen has been used for 3D in vitro pharmacological testing (Magdeldin et al., 2014) or cell invasion (Nyga et al., 2013) with HT29 and HCT116 cell lines, as well as to stimulate morphological differentiation in some colon cell lines (Del et al., 1991). Collagen hydrogel cultures have also been utilized to explore the role of matrix-degrading metalloproteinases that are expressed in colorectal cells (Yamamoto et al., 2008). Typically, epithelial and mesenchymal cells interact with type IV collagen via receptors such as integrins. HA have also been found to be abundant in the ECM of CRC. Lue et al. used hydrogelsmade of hyaluronan and collagen I, to create a 3D matrix for the co-culture of CRC PDO and cancer associated fibroblasts (CAFs) (Luo et al., 2021). They discovered that these hydrogels could retain the critical molecular properties of the original patient tumors in CRC PDOs. Digging into the cellular compartment of TME is the next step after exploring its acellular components.

3.2 Stroma cells in the tumor microenvironment

It is well-known that the TME of CRC is composed of both malignant and various non-cancerous cells. Stromal cells are a key component of the non-cancerous cells in the TME. It consists of several cell types including mesenchymal stromal cells (MSCs), fibroblasts, and endothelial cells (Figure 4). As a crucial part of the TME, the tumor stroma influences and contributes to cancer progression, metastasis and treatment resistance.

Figure 4
www.frontiersin.org

Figure 4. Stromal cell compartment is a complex key environment surrounding the cancer cells, consisting of MSCs, fibroblasts, and endothelial cells. Created with BioRender.com.

3.2.1 Mesenchymal cells

In advanced CRC, the stroma contributes up to 50% of the primary tumor mass (Binnewies et al., 2018), with the majority of stromal cells being MSCs. A number of mesenchymal-derived cell types with similar characteristics are included, such as myofibroblasts and CAFs, and they can be resident cells or recruited to the tumor site to support development (Sai et al., 2019). CRC cells cultured in 3D using Matrigel with combinations of crypt growth factors such as EGF, Wnt, R-Spondin 1 and Noggin formed an invasive disc-like colonies (Ludwig et al., 2013). 3D CRC cultures remain responsive to growth factor stimulation and the invasive phenotype is reversible. CRC cells lose the epithelial phenotype and simultaneously gain the mesenchymal phenotype, undergoing epithelial-to-mesenchymal transition (EMT) that allows them to metastasize (Buhrmann et al., 2019). Furthermore, the TME were shown to influence the EMT of CRC cell lines when grown in 3D-alginate culture (Buhrmann et al., 2021). Indeed, EMT is a critical process in cancer metastasis which enhanced their invasiveness and mobility.

Moreover, CRC cells co-cultured with mesenchymal stromal cells and immune cells showed that the presence of mesenchymal cells in the TME promote an immunosuppressive environment (Leonard et al., 2021). Leonard et al. developed a 3D model of CRC that includes MSCs and immune cells to better mimic the TME and study its effects on both tumor growth and treatment response (Leonard et al., 2021). They used a gelatin-methacryloyl-based hydrogel culture system incorporating CRC cells, MSCs, and a monocyte cell line. The presence of stromal cells increased the transcription of matrix remodeling proteins (FN1 and MMP9) and the release of tumor-promoting immune molecules such as macrophage inhibitory factor (MIF), Serpin E1, chemokine (C-X-C motif) ligand 1 (CXCL1), interleukin-8 (IL-8) and chemokine (C-X-C motif) ligand 12 (CXCL12). Stromal cells altered the expression of immunotherapeutic targets on cancer cells, such as epidermal growth factor receptor (EGFR), Cluster of Differentiation 47 (CD47) and Programmed death-ligand 1 (PD-L1). Treatment with an EGFR inhibitor (PD153035) showed altered PD-L1 expression in CRC cells, but only in the absence of MSCs.

3.2.2 Cancer-associated fibroblasts

CRC development involves dynamic interaction among malignant cells, components of the microenvironment (stromal and vascular endothelial cells), and the immune system (Mascaux et al., 2019). Several cell types from the TME can be recruited in the cancer progression and become recruited to fulfill critical pro-tumorigenic roles. This enables new cancer cells to elude immune system identification and establish a niche in which to grow unchecked (de Visser and Joyce, 2023). Fibroblasts are among the first cell types recruited within the TME, and they undergo a process known as activation when the crosstalk with cancer cells begins. Studies using 3D interpenetrating networks of collagen and alginate have shown that CAFs can switch between inflammatory and myofibroblastic states, which affect their role in tumor progression (Cao et al., 2021). The ECM is mainly produced and organized by fibroblasts, which serve as a structural scaffold for tissue architecture as well as a reservoir for growth factors and cytokines that can be released during matrix remodeling and cleavage (Arroyo and Iruela-Arispe, 2010). These cytokines and growth factors can influence the onset and course of CRC (Crotti et al., 2017). High stromal infiltration has been shown to be related to shorter overall survival in CRC patients. This is because both MSCs and CAFs secrete growth factors such as fibroblast growth factor (FGF) or hepatocyte growth factor, which can promote tumor growth (Turley et al., 2015), as well as TGF-β which fosters metastasis and leads to an immunosuppressive environment (Tauriello et al., 2018). Cao et al. showed the dual roles of CAFs in promoting and inhibiting tumor growth by using a 3D interpenetrating network of collagen and alginate (Cao et al., 2021). They found that CAFs can switch between the inflammatory-state having an anti-tumorigenic effect to the EMT-promoting myofibroblastic-state depending on the mechanical properties of the 3D environment. Understanding the conditions that cause CAFs to switch states can help in developing targeted therapies for cancer treatment by either promoting the anti-tumorigenic properties or inhibiting the pro-tumorigenic functions. Micalet et al. investigated how CAFs from different CRC patients affect the TME, particularly focusing on tissue stiffness (Micalet et al., 2024). Different patient-derived CAFs showed varied abilities to remodel the ECM where some stiffened the matrix through active contraction, while others softened it via enzymatic activity.

3.2.3 Endothelial cells

Cell heterogeneity within a 3D model is an advanced concept for evaluating drug efficacy, understanding chemoresistance and developing TME-targeted therapies. In one study, a 3D multicellular model for CRC was constructed using the CRC cell line SW480 with CAFs and endothelial cells, showing improved expression of several tumor-related genes including IL1B, FCGR2A, FCGR3A, CYBB, SPI1, CCL2, ITGAM, and ITGB2 (Wang et al., 2023). Other studies have reported increased drug resistance when co-culturing CRC cell lines with CAFs and endothelial cells (Zoetemelk et al., 2019). Briefly, they used HCT116, SW620 and DLD1 cells, which were co-cultured with fibroblasts and endothelial cells in 3D spheroids. Treatment was performed with 5-fluorouracil (5−FU), regorafenib and erlotinib. The results showed a dose-dependent increase of erlotinib sensitivity. Interestingly, simple 3D spheroid and 3D co-cultures responded distinctly to drug treatment, and the signalling pathways were also regulated differently. Moreover, Chen et al. have developed a 3D printed in vitro model which mimics the TME by co-culturing CRC cell lines, CAFs, and tumor-associated endothelial cells in 3D-printed scaffolds (Chen et al., 2020). This 3D model exhibited physiological activity with high drug resistance. To preserve the complex TME of CRC, freshly excised CRC samples were fragmented and placed between collagen type I sponges in a “sandwich-like” format and then cultured in a perfusion bioreactor (Manfredonia et al., 2019). This approach shows promise for more accurate in vitro models which can predict patients’ responses to treatments, potentially guiding therapeutic decisions more effectively. Therefore, compared with a single-cell model, the use of a 3D multicellular co-culture models is indeed a better option for mimicking the CRC environment.

3.3 Immune cells in the tumor microenvironment

Immune cells are additional major cell types following stromal cells in the TME. The immune infiltrate in CRC varies by subtype and may include T cells, B cells, neutrophils, monocytes, macrophages, natural killer (NK) cells and mast cells (Zhang et al., 2018). Under normal conditions, the immune system can eliminate cancer cells. Tumors have devised strategies to overcome this by polarizing immune cells into tumor-promoting phenotypes (Heichler et al., 2020).

Dynamic and heterogeneous interactions between tumor cells and the surrounding microenvironment fuel the occurrence, progression, invasion, and metastasis of solid tumors. The TME plays a key role in modifying the plasticity of tumor cells and offers immunological escape strategies. Tumor-associated macrophages (TAMs) play a pivotal role in this process (Figure 5). Numerous studies have shown that TAMs can cause genetic instability in cancer cells (Bonavita et al., 2015), induce angiogenesis, increase tumor development and contribute to ECM degradation (Guerriero, 2018). TAMs can differentiate into pro-inflammatory anti-tumorigenic M1 and anti-inflammatory, pro-tumorigenic M2 phenotype. M1 are stimulated by TNF-α and IFN-γ to produce pro-inflammatory cytokines such as IL-1, IL-6, IL-12, IL-23, CXCL-10 and TNFα and in that way contribute to destroying tumor cells in the TME. The M2 phenotypes are, on the other hand, stimulated by IL-4 and IL-13 to produce and secrete growth factors such as VEGF and TGF-β, MMPs and other cytokines such as IL-10, IL-13 and IL-4, and in that way contribute to increased proliferation of tumor cells and angiogenesis in the TME. Depending on which chemokines, cytokines and growth factors are present in the TME these two types of macrophages can transform into each other, which is important for CRC progression and metastasis (Hou et al., 2024). For instance, STAT3 has been shown to be the main transcription factor for TAM transformation (Xia et al., 2023) and inhibition of the STAT3 signaling leads to reduced CRC metastasis (Zhang et al., 2022).

Figure 5
www.frontiersin.org

Figure 5. Interaction between CRC and TAMs. Different stimuli can polarize macrophages into two basic types, anti-tumorigenic M1 and pro-tumorigenic M2 phenotypes. M1 macrophages can be stimulated by interferon IFN-γ and express inflammatory factors including interleukin IL-1β, IL-6, and tumor necrosis factor TNF-α. M2 macrophages are stimulated by IL-4 and IL-13 and express IL-10 and transforming growth factor TGF-β, among other cytokines. Created with BioRender.com.

Immune cells in the TME are represented by the diversity of cells including TAMs, dendritic cells, regulatory T cells (Tregs), cytotoxic T cells (CD8+ T cells), and myeloid-derived suppressor cells (MDSCs) (Joyce and Pollard, 2009). Depending on the tissue context and the presence of signaling molecules, these cells could either promote or suppress tumor progression, contributing to immune modulation in cancer. A study of the TME from left-sided colon cancer and right-sided colon cancer based on scores from TCGA revealed that there was a difference in TME profiles (Liu D. et al., 2024). Left-sided colon cancer had high scores of non-differentiated M0 macrophages, activated CD4+ T cells, dendritic cells, NK cells and monocytes. On the other hand, right-sided colon cancer had higher scores of inflammatory M1 macrophages, neutrophils and CD8+ T cells resulting in a worse prognosis, especially for advanced stage (III/IV) colon cancer. Right-sided colon cancer showed elevated expression of programmed cell death protein-1 (PD-1), suggesting that these patients might respond better to treatments with immune checkpoint inhibitors (Liu D. et al., 2024). Numerous studies have shown that TAMs can cause genic instability in cancer cells (Bonavita et al., 2015), induce angiogenesis, increase tumor development and contribute to ECM degradation (Guerriero, 2018).

TAMs also produce EGF which can increase cancer cell migration and invasion by binding to EGFR expressed on cancer cells (Zhao et al., 2016). Macrophages are involved in regulation of collagen fibers and might thus promote cancer migration towards blood vessels since cancer cells use collagen for migration (Joyce and Pollard, 2009). PD-L1 is expressed by cancer cells, dendritic cells, B cells as well as TAMs, indicating pro-tumorigenic function which complicates clarification of the mechanism behind the anti-PD-L1 therapies (Du et al., 2024; Leonard et al., 2024). Strasser et al. showed that the TME of CRC consists of both pro- and anti-inflammatory immune cells which promotes progression of the CRC and might explain why inhibition of PD-1/PD-L1 is not that effective in CRC (Strasser et al., 2019). Three CRC cell line-based models of HCT-116, an in vitro 2D model, an in vitro 3D model of spheroids and an in vivo xenograft mice model were used to test the effect of sequential micro-immunotherapy medicine on M1 and M2 macrophages. This study suggests that 3D spheroid models provide better understanding and analysis of drug response (Jacques et al., 2022).

Tregs are either circulating or tissue-localized cells expressing a variety of inhibitory proteins that serve as control of immune safety. With other cells present in TME such as M2 macrophages, tolerogenic dendritic cells, and myeloid-derived suppressor cells, Treg cells can maintain the immune equilibrium by controlling function of different T cells by expressing a variety of proteins such as PD-1, lymphocyte activation gene-3 (LAG3) and cytotoxic T-lymphocyte antigen-4 (CTLA-4). Tregs are induced and differentiated by traditional T cells and are suggested to have the ability to inhibit activation and differentiation of CD4+ helper T cells and CD8+ cytotoxic T cells. However, Tregs also appear to stop the host´s anti-tumor response and enhance tumor immune escape, growth and metastasis (Verma et al., 2022; Li C. et al., 2020). This pro-tumor property of Tregs is due to the secretion of molecules (TGF-β, IL-10, and IL-35) which regulate the expression of inhibitory receptors. Since Tregs cannot be eliminated from the tissue, approaches to controlling actions of Treg cells have been proposed in form of immune checkpoint inhibitors (Li C. et al., 2020). Since immunotherapies still fail to gain CRC survival, new functional assays and cancer cell models are required. Natural killer (NK) cells belong to innate lymphoid cells and have the capacity to kill tumor cells by releasing cytotoxic molecules and by producing cytokines and chemokines that recruit other immune cells such as dendritic cells (Marchalot and Mjösberg, 2022). However, Mao et al. showed that NK cells exhibited tumor-activating characteristics when they were co-cultured with tumor cells thus promoting progression of colon cancer liver metastasis (Mao et al., 2024). Courau et al. have showed that co-culture of CRC cell lines (HT-29 and DLD1) together with T and NK cells in a 3D spheroid model is a good model of studying tumor-lymphocyte interactions and immunotherapy (Courau et al., 2019).

4 Models for treatment of CRC and mCRC

Preclinical cancer research entails the testing of novel drugs in non-human subjects, necessitating several experiments based mostly on 2D-cultured cancer cell lines and animal experiments. For decades, in vivo models have been regarded as the gold standard for oncological research to investigate the complex multicellular components of tumors (Tosca et al., 2023).

The research on the TME has recently been gaining attention due to its important role in tumor growth, progression, and response to therapy. Because of this, the development of 3D in vitro cancer models that mimic the interactions in the TME, the tumor structure and complexity is of great relevance for drug development. The antimetabolite 5-FU remains a crucial treatment strategy for CRC and the use in combination with folinic acid was shown to be a better outcome for patients (Petrioli et al., 1995). The incorporation of oxaliplatin has shown additional survival benefits (Wolpin et al., 2007). In metastatic disease, treatment strategies have become more complex, involving various combinations of chemotherapy FOLFIRI, (fluorouracil, folinic acid and irinotecan); FOLFOX, (fluorouracil, folinic acid and oxaliplatin); FOLFOXIRI, (fluorouracil, folinic acid, oxaliplatin and irinotecan) and XELOX, (capecitabine plus oxaliplatin). Results of molecular research have demonstrated the need to profile each mCRC for different mutations. Targeted therapies have further expanded the treatment options, but novel therapies still need to be developed (Aparicio et al., 2020). Multiple clinical trials have shown that tumors with activating mutation do not benefit from targeted therapies (Aparicio et al., 2020).

Even though survival rates have increased with the introduction of targeted therapies, one of the major challenges in treating CRC is the development of treatment resistance, especially in metastatic cases. CRC is a genetically diverse disease and new treatment strategies are needed, such as improved targeted therapies and immunotherapy. Immunotherapy is clinically used and FDA-approved for patients with high MSI, MSI-H, or mismatch repair-deficient (dMMR) CRC. These tumors have numerous mutations that make them more visible to the immune system. The dMMR/MSI-H cancers are associated with strong lymphocytic infiltration in and around the tumor (André et al., 2020). Immune checkpoint inhibitors (ICIs) have as a consequence emerged as a promising immunotherapy for CRC patients with this type of mutations (Passardi et al., 2017; Hirano et al., 2021). These therapies aim to enhance the immune system’s ability to attack tumor cells by blocking the proteins responsible for inhibiting the immune response, such as PD-1 and CTLA-4. However, microsatellite stable (MSS) CRC, which represents the majority of cases, has shown limited response to ICIs since MSS tumors often have low levels of lymphocytic infiltration and low levels of immune checkpoint molecules (Chen et al., 2021). Ongoing research focuses on combining ICIs with other immunotherapies or conventional treatments to enhance efficacy in MSS CRC (Makaremi et al., 2021).

Cell growth in 3D settings not only promotes phenotypic changes in cell morphology, sensitivity to stimuli, cell activities and gene expression patterns, but it also affects the response to therapeutic agents (Romero-López et al., 2017). Indeed, both normal and cancerous cells retain their distinct behaviors in the body owing to their 3D environment, which includes heterogeneous and dynamic cell-cell and cell-matrix interactions (Kessenbrock et al., 2010). Hachey et al. showed that when CRC cell gene expression, heterogeneity, growth, and responsiveness to standard chemotherapy were examined, cells growing in vascular micro-tumors (VMT) more closely resembled those growing as in vivo murine xenografts (Hachey et al., 2021). It is likely that 3D cell culture models have demonstrated exceptional capabilities in bridging the gap between oversimplified 2D systems and subsequent animal experiments for safety assessment prior to clinical trials. Increasing the predictive capacity of 3D culture models may help to minimize the number of animals used by the pharmaceutical industry for drug efficacy and toxicity testing. 3D cell cultures also demonstrated stronger stability and longer life spans than 2D cell cultures did, making them better suited for long-term studies on cell interactions or drug effects. Similarly, the development of patient-derived cells in such 3D cultures may provide unparalleled opportunities for personalized pharmacogenetic techniques that animal models cannot address.

By using Kras mutant mouse models of CRC, it was reported that wild-type Kras plays a substantial role in the fitness, evolution, and treatment susceptibility of Kras mutant cells in vivo (Najumudeen et al., 2024). The combination chemotherapy regimen based on oxaliplatin remains the first-line treatment for CRC, notably including the FOLFOX regimen and the XELOX regimen. However, CRC becomes resistant to oxaliplatin, and the underlying mechanism is unknown (Deng et al., 2024). Deng et al. established a CRC subcutaneous tumor model and metastasis model via oxaliplatin induction to generate human CRC drug-resistant cell lines (Deng et al., 2024). They demonstrated that increased H3K79 methylation may be responsible for the resistance of CRC cells to oxaliplatin.

Inhibiting HIF-1α can improve the CRC microenvironments and increase treatment sensitivity (Su et al., 2024). Knocking down HIF-1α or its upstream regulator BIRC2 in animal experiments leads to tumor growth inhibition by increasing CD8+ T cell infiltration. Moreover, bleomycin or doxorubicin had greater anti-tumor effect in vivo when HIF-1α was knocked down. Notably, the TGF-β produced by cancer cells and CAFs is central to immune suppression within the TME and contributes to tumor immune evasion and poor responses to cancer immuno-therapy (Batlle and Massagué, 2019). TGF-β released in the TME attracts fibroblasts and induces the production of CAFs (Lebrun, 2012). Additionally, TGF-β promotes immune evasion in genetically reconstituted mCRC (Tauriello et al., 2018). In mCRC mouse models, Li et al. detected a decrease in CAFs through the TGF-β receptor inhibitor SB525334, which sensitized mCRC to immunotherapy by improving the tumor immune microenvironment and led to significantly reduced tumor uptakes of 68Ga-FAPI by PET/CT (Li K. et al., 2024). The 68Ga-FAPI PET/CT imaging helps select patients with mCRC who can benefit from immunotherapy and guides the precise timing of TGF-β inhibition to optimize the combination strategy with immunotherapy.

Many studies illustrated the efficiency of 3D in vitro models to replace animal models in CRC research. Vlachogiannis et al. developed a biobank of PDOs from metastatic, heavily-pretreated colorectal and gastroesophageal cancer patients, to assess molecular profiles and drug responses. PDOs were derived from sequential biopsies taken at different stages including baseline, at the time of best response and at disease progression, as well as from multiple tumor sites. They tested 19 PDOs in a screening using a library of 55 drugs currently in phase I-III clinical trials or in clinical practice. By comparing the drug responses of PDOs and PDO-based orthotopic mouse tumor xenografts with patient responses in clinical trials, they concluded that PDOs effectively reflect patient heterogeneity and can be a valuable tool for predicting the response of specific therapies. It was also proved by Svanström et al. that the environment in a 3D culture model more closely resembles the native tissue. They showed that cells cultivated in both patient-derived and 3D printed scaffolds responded similarly to hypoxic circumstances, which is a significant feature in tumors. Moreover, Chen et al. co-cultured CRC and CAFs in 3D printed scaffolds to mimic ECM. The model showed a physiological activity similar to in vivo tumors, with high drug resistance and overexpression of tumor-related markers.

The patient-derived xenograft (PDX) model is an effective tool for studying tumors because it can maintain 3D cell-cell interactions and capture tumor heterogeneity (Drost and Clevers, 2018). Tumor-derived organoids in culture may accurately reflect the gene expression patterns, gene regulatory networks, point mutations, and DNA methylation patterns of tumor cells in vivo (Wang et al., 2022). Normal-tissue-derived organoids retain the normal genomic features of normal epithelial cells in vivo. Recently, established 3D organoid culture technologies have transformed cancer research by enabling more realistic and scalable replications of both tumor and microenvironmental structures (Drost and Clevers, 2018; Tuveson and Clevers, 2019). It can serve as an appropriate preclinical model for the evaluation of high-throughput drug response screening and the mechanistic studies of tumorigenesis, invasiveness, and metastasis (van de Wetering et al., 2015). Organoid-based 3D models have been proposed as a bridge between in vitro and in vivo models (Kim et al., 2020). Additionally, the CRC organoid orthotopic transplantation technique and a genetically engineered autochthonous mouse adenoma model were used to conduct a comprehensive epigenomic and transcriptomic analyses to determine the variables required for tumor initiation in these cancers (Goto et al., 2024). This study investigated how naive colon cancer organoids produced in vitro with Apc-null, KrasG12D and Trp53-null (AKP) mutations acclimated to the in vivo native colonic environment. These findings revealed that SOX17, a transcription factor involved in endoderm and fetal foregut development (Spence et al., 2009), is required for the growth of adenomas and CRC. Importantly, organoid models have limitations due to closed cystic structure instead of an in vivo-like apically open architecture (Sato et al., 2009), short lifespan that requires breaking up the culture every few days for passaging (Nikolaev et al., 2020), and lack of topobiological stability and consistency owing to stochastic growth in 3D matrices (Sato et al., 2009).

5 Nanotools for diagnostic and therapeutic applications of 3D models and their ECM components

Several excellent reviews focusing on the nano- and microengineering of the TME for different diseases already exist (Jin et al., 2020; Kim et al., 2023; Chen et al., 2018; Song et al., 2018). Cancer diagnostics and therapeutics can improve significantly by using nanotools for imaging and treatment applications (Jin et al., 2020). The collection of nanoscale tools discussed in a review by Jin et al. includes quantum dots, nanoshells, gold nanoparticles (NP), liposomes, carbon nanotubes, polymeric micelles and dendrimers. Moreover, recently described engineered therapeutic models involve a variety of 3D model scaffold systems such as electrospun nanofibers, nanoprinted scaffolds, self-assembled peptide hydrogels, and systems incorporating dynamic physiological features into both nano- and microstructure substrates for cancer research (Kim et al., 2023; Chen et al., 2018). Electrospun nanofibers with the size of collagen fibrils, or surface modified with ECM-derived proteins were preferable (Chen et al., 2018). Drug screening platforms using micro- or nanoengineered technologies are in clinical use. Patient-derived tissue slices on microfluidic devices were tested for multiplexed drugs in both glioblastoma and metastatic CRC. Referring to clinical trials, Kim et al. mention that patient-derived organoids-on-a-chip were tested to screen drugs and assessing response to guide CRC treatment of the patients (Kim et al., 2023; Li et al., 2011). Moreover, tissues have been employed to establish a 3D bio-printed model and to predict clinical efficacy of chemotherapeutic drugs of CRC patients. Song et al. describe advanced modern biomicroscopic tools with commonly used methods and materials to engineer 3-D cancer or other disease models (Song et al., 2018). A fabricated 3D cell-mimicking hydrogel microtissue with a photocurable hydrogel as the ECM could control the cell size as well as the cell type (Li et al., 2011). In another example, a method to encapsulate single cells within 3D matrix structures using photopolymerization technique in hydrogel micro-niches was described to allow both cell adhesion and nutritional permeability (Bao et al., 2017). A study by Yan et al. revealed that a novel supramolecular up-conversion NP with incorporated platinum (IV) oxaliplatin prodrug and a polymer with inhibition capacity for a certain bacteria existing in the community of CRC, as well as with polyethylene glycol-azobenzene could together enhance drug-loading and enable on-demand drug release for drug-resistant CRC treatment (Yan et al., 2024). This study indicates that multiple dynamic chemical designs integrating drug loading and release of a single system provide a promising candidate for precision therapy.

Preclinical studies indicate that TAMs represent an attractive target for cancer therapeutics (Miao and Huang, 2015). TAMs express high amounts of the mannose receptor, which has been used as a targeting ligand for NP-based TAMs delivery (Zhu et al., 2013). With better knowledge about M1 macrophages having antitumorigenic properties, focus of NP development has shifted from exclusively depleting and imaging all TAMs to modulating the ratio of M1/M2 macrophages for improved therapy (Miao and Huang, 2015). CAFs secrete diverse growth factors, cytokines, and extracellular matrix (ECM) proteins that fuel tumor growth and progression. To specifically inhibit CAF activation, proliferation, or function, small-molecule inhibitors, antibodies, or NPs can be tailored for therapeutics (Shah et al., 2025). However, considerations must be taken on any potential adverse effects on normal fibroblasts.

5.1 Modulating the stroma of the tumor

The modulation of stromal cellular components such as endothelial cells, CAFs or TAMs, by small molecules or nanodrugs can facilitate the remodeling of tumor blood vessels or ECM, are recognized as major components that promote cancer progression, therapy resistance, and metastasis formation. Miao et al. highlight in their review that this can be performed by distribution of NPs to these cellular components in stroma (Miao and Huang, 2015). Major components of type IV collagen, laminin, entactin (nidogen) and fibronectin are building up the vasculature basement membrane. The higher tumor vasculature compared to healthy vasculature leads to leaky and loosely compacted vasculature, which tends to be abnormally permeable to macromolecules and NP (10–100 nm, in diameter). Stroma cells, on the other hand, can compress vessels and inhibit NP penetration. To improve drug penetration, proteases including hyaluronidase, collagenase, matrix MMP-1 and MMP-8 are frequently used to decrease the level of NP-limiting tumor glycosaminoglycans (Miao and Huang, 2015). Another approach reviewed by Miao et al. was depleting HA by using PEGylated human recombinant PH20 hyaluronidase (PEGPH20). This was shown to increase macromolecule permeability and augment chemotherapy responses in a pancreatic model. Inhibiting the enzyme critical for the stabilization of collagen networks, LOX, is another strategy (Liu et al., 2025). Findings highlighting the challenge of targeting ECM synthesis in cancer therapy are for example, studies using the combination of the LOXL2 antibody Simtuzumab with FOLFIRI chemotherapy or gemcitabine, which did not show significant clinical benefit in 280 patients with pancreatic cancer or CRC as reviewed by Liu et al. (2025).

6 Discussion

The use of animal models is both economically and ethically costly. Thus, there is the universal commitment that animal use in research should be reduced, replaced, and refined, known as the 3R principle established by Russell and Burch in 1959. According to the 3R principle, animal experiments should be replaced with alternatives whenever possible. Additionally, irreproducibility in scientific research has become a major issue. Despite the need for rigor when writing a scientific paper, more than half of publications are regarded as non-reproducible. In the context of animal research, selecting an appropriate experimental model within the 3R concept is crucial, as irreproducibility causes an annual loss of 28 billion dollars in biomedical research without fruitful outcomes. The expansion of the 3R to the 5R, by introducing “robustness” and “reproducibility”, may provide a framework for research improvement. There is a significant lack of data specifications in scientific articles, revealing gaps in the presentation of critical details, such as environmental conditions and experimental protocols. Certainly, one of the advantages of 3D models is their ability to simulate the biological TME in the laboratory. From this perspective, reproducibility is extremely beneficial, as is the ability to standardize experiments.

Soon, the utilization of 3D in vitro cancer models may become a required step between 2D in vitro and in vivo animal models. Identifying and eliminating those drugs that have no interesting efficacy in 3D in vitro cultures will reduce animal use as well as the associated costs and ethical issues. This could increase the number of effective drug candidates that have advanced to clinical development, thereby lowering the number of patients who receive ineffective treatments and increasing the success rate of clinical trials.

Additionally, in vivo model has several technical constraints such as the number of animals required for time-point experiments and the sensitivity of cell imaging in vivo. Another drawback is that species differences may impair the clinical translation of results. Moreover, animal models are both time-consuming and require expensive commitments, not to mention the ethical concerns. The need for precise and trustworthy results has driven the development of more biomimetic and clinically relevant disease models, to prevent early clinical trials and save time and resources. Various in vitro models have been published and used to assess potential drug candidates, which are often examined in 2D cell monocultures. Only 5% of drugs proven to be active in such cell culture models have reached the clinical trials (Kola and Landis, 2004; Ibarrola-Villava et al., 2018). Current evidence shows that 3D culture models may better capture the mechanisms of drug resistance identified in tumors. 3D in vitro models can be very efficient in term of multiple drugs evaluation as shown in the study by Kondo et al. with a large-scale screening of 2,427 compounds on CRC organoids to uncover numerous “hit” drugs (Kondo et al., 2019). To solve the time-consuming process of PDO derivation and expansion, Ding et al. developed an automated microfluidics droplet platform that can construct patient-derived microorganospheres (MOS), allowing for quick clinical high-throughput drug selection in a substantially shorter period (Ding et al., 2022). By analysing samples from eight metastatic CRC patients, the authors revealed that studies using MOS can provide results within 7–14 days of biopsy collection, with drug responses consistent with clinical outcomes. Notably, the original tumor’s stromal and immune cells were maintained in the MOS, making it an effective tool for testing immunotherapies.

The advances in understanding cancer disease mechanisms related to TME will lead to a major revolution in the research field. However, the magnitude of this impact is determined by the effectiveness with which biomedical research breakthroughs are converted into clinical practice. This translation process entails the development of new drugs, of medical equipment, and of clinical procedures. According to National Institutes of Health (NIH), bringing a new drug or medical device from development to market takes on average 14 years and $2 billion (Health NIo, 2015). Therefore, we concur that laboratory results from 3D CRC models have a long way to go to clinical use. The challenges to translating those findings into clinical practice are well documented for each 3D approach (Bose et al., 2022; Price et al., 2023; Peng et al., 2025).

3D in vitro models of CRC own numerous potentials for guiding clinical decisions through several key mechanisms. Like in personalized drug screening, PDO 3D models can be generated immediately from tumor biopsies. These models keep the original tumor’s genetic, molecular, and histological properties, allowing clinicians to evaluate multiple therapeutic alternatives in vitro before deciding the most effective regimen for the patient (Thorel et al., 2024). This personalized approach can reduce trial-and-error prescribing while improving outcomes by identifying treatments that are more likely to succeed. In prediction of drug resistance, 3D models that can simulate the complexity of CRC TME, including ECM stiffness and cell heterogeneity, are valuable factors since those are known to influence drug resistance in CRC (Liu et al., 2020). Understanding how cancer cells interact with these factors in a controlled 3D environment helps clinicians to anticipate resistance mechanisms and adjust therapy accordingly, potentially combining treatments to overcome resistance. Additionally, while determining whether a drug works, 3D in vitro models enable researchers and clinicians to test different dosing regimens, combinations, and sequences of therapies in a milieu that mimics tumor physiology. This provides more information about treatment strategies, potentially reducing side effects and increasing efficacy. Furthermore, the benefit of quick testing in clinical timeframes since advances in bioengineering and culture techniques have reduced the time required to grow and test PDO 3D models, making it increasingly feasible to incorporate results into clinical timelines. For example, mini-organoids can sometimes be tested within 1–2 weeks, offering relevant data for oncologists treating aggressive or late-stage CRC (Zhou et al., 2021).

7 Conclusion

3D in vitro model systems that mimic the complex nature of the TME allow studies of cellular and molecular interactions offering valuable insights that would be challenging to obtain solely from clinical samples alone. There have been recent advancements in spheroid models and PDOs enhancing the understanding of the heterogeneic physiology of the TME. Emphasis on systematically including stromal cells, immune cells and endothelial cells in the 3D culture models, while analyzing these systems under defined experimental conditions will further improve the testing the effects of therapeutic agents and reliable outcome.

This review highlights the need for of validated in vitro models in cancer research. Today’s animal testing must be substituted with standardized 3D models including both cellular and acellular ECM. Establishing a 3D in vitro model in the cancer research area which considers the TME condition as much as possible could turn this fiction into reality. Furthermore, recent improvements in 3D culture design and co-culture techniques, will pave the way for the creation of real TME model that includes an increasing number of cell types. Combining these models with biological scaffold and ethical animal-free system is what we suggest for the future of CRC 3D in vitro models.

Recent advancements in technology are paving the way for the development of high-throughput 3D tumor model screening platforms. Combining multiple treatment modalities and nanotechnology that target different components of the TME holds promise for improving therapeutic outcomes. Multiplexed immunofluorescence and spatial transcriptomics are examples of novel methodologies that reveal detailed insights in the cellular composition and spatial organization of the TME. These approaches enable the identification of unique TME signatures associated with therapy response and resistance, guiding the development of personalized treatment strategies.

While 3D models could offer considerable advantages as previously discussed, their practical application in clinical settings remains limited. Researchers encounter several hurdles in bridging the gap between laboratory findings and real-world clinical use. One challenge is the lack of model standardization, as there is currently no universally agreed protocol to generate 3D in vitro models. Therefore, variability might lead to inconsistent data across laboratories. This lack of uniformity hinders reproducibility and complicates cross-study comparisons, which are essential for clinical validation. Moreover, not all patient tumors yield viable organoids, particularly in cases of advanced CRC or treatment-resistant CRC.

Nevertheless, even when reliable data is collected from 3D models, incorporating it into existing clinical decision-making pathways is difficult. Model-derived knowledge must be included into the decision-support system; otherwise, valuable laboratory findings remain underutilized. To increase the clinical utility of CRC 3D in vitro models, international guidelines or consensus protocols for culturing and analyzing 3D models must be in place. Encouraging open-source platforms for sharing validated techniques, including materials used, culture periods, and response evaluation measures, will benefit both researchers and clinicians. In addition, miniaturized assay formats (e.g., 96- well plate) optimized for 3D models could reduce reagent costs and increase throughput. Finally, training clinicians via interdisciplinary workshops and continuing medical education courses in precision oncology and 3D model interpretation might be a possible way.

Author contributions

VH: Conceptualization, Software, Writing – original draft, Writing – review and editing. ZP: Conceptualization, Writing – original draft. MH: Conceptualization, Writing – original draft, Writing – review and editing. HT: Investigation, Writing – original draft. AW: Conceptualization, Investigation, Writing – original draft, Writing – review and editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. Knowledge Foundation, Sweden. Project #20220218 and EU REA project REVERT #844098.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Abbas, Z. N., Al-Saffar, A. Z., Jasim, S. M., and Sulaiman, G. M. (2023). Comparative analysis between 2D and 3D colorectal cancer culture models for insights into cellular morphological and transcriptomic variations. Sci. Rep. 13 (1), 18380. doi:10.1038/s41598-023-45144-w

PubMed Abstract | CrossRef Full Text | Google Scholar

Ahmad Zawawi, S. S., Salleh, E. A., and Musa, M. (2024). Spheroids and organoids derived from colorectal cancer as tools for in vitro drug screening. Explor Target Antitumor Ther. 5 (2), 409–431. doi:10.37349/etat.2024.00226

PubMed Abstract | CrossRef Full Text | Google Scholar

Alzahrani, S. M., Al Doghaither, H. A., and Al-Ghafari, A. B. (2021). General insight into cancer: an overview of colorectal cancer (Review). Mol. Clin. Oncol. 15 (6), 271. doi:10.3892/mco.2021.2433

PubMed Abstract | CrossRef Full Text | Google Scholar

André, T., Shiu, K. K., Kim, T. W., Jensen, B. V., Jensen, L. H., Punt, C., et al. (2020). Pembrolizumab in microsatellite-instability-high advanced colorectal cancer. N. Engl. J. Med. 383 (23), 2207–2218. doi:10.1056/nejmoa2017699

PubMed Abstract | CrossRef Full Text | Google Scholar

Aparicio, J., Esposito, F., Serrano, S., Falco, E., Escudero, P., Ruiz-Casado, A., et al. (2020). Metastatic colorectal cancer. First line therapy for unresectable disease. J. Clin. Med. 9 (12), 3889. doi:10.3390/jcm9123889

PubMed Abstract | CrossRef Full Text | Google Scholar

Arroyo, A. G., and Iruela-Arispe, M. L. (2010). Extracellular matrix, inflammation, and the angiogenic response. Cardiovasc Res. 86 (2), 226–235. doi:10.1093/cvr/cvq049

PubMed Abstract | CrossRef Full Text | Google Scholar

Aumailley, M. (2013). The laminin family. Cell Adhesion and Migr. 7 (1), 48–55. doi:10.4161/cam.22826

PubMed Abstract | CrossRef Full Text | Google Scholar

Bao, M., Xie, J., Piruska, A., and Huck, W. T. S. (2017). 3D microniches reveal the importance of cell size and shape. Nat. Commun. 8 (1), 1962. doi:10.1038/s41467-017-02163-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Batlle, E., and Massagué, J. (2019). Transforming growth factor-β signaling in immunity and cancer. Immunity 50 (4), 924–940. doi:10.1016/j.immuni.2019.03.024

PubMed Abstract | CrossRef Full Text | Google Scholar

Berg, K. C. G., Eide, P. W., Eilertsen, I. A., Johannessen, B., Bruun, J., Danielsen, S. A., et al. (2017). Multi-omics of 34 colorectal cancer cell lines - a resource for biomedical studies. Mol. Cancer 16 (1), 116. doi:10.1186/s12943-017-0691-y

PubMed Abstract | CrossRef Full Text | Google Scholar

Binnewies, M., Roberts, E. W., Kersten, K., Chan, V., Fearon, D. F., Merad, M., et al. (2018). Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat. Med. 24 (5), 541–550. doi:10.1038/s41591-018-0014-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Bonavita, E., Galdiero, M. R., Jaillon, S., and Mantovani, A. (2015). “Phagocytes as corrupted policemen in cancer-related inflammation,”. Advances in cancer research. Editors X.-Y. Wang, and P. B. Fisher (Academic Press), 128, 141–171. doi:10.1016/bs.acr.2015.04.013

CrossRef Full Text | Google Scholar

Bonnans, C., Chou, J., and Werb, Z. (2014). Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 15 (12), 786–801. doi:10.1038/nrm3904

PubMed Abstract | CrossRef Full Text | Google Scholar

Boot, A., van Eendenburg, J., Crobach, S., Ruano, D., Speetjens, F., Calame, J., et al. (2016). Characterization of novel low passage primary and metastatic colorectal cancer cell lines. Oncotarget 7 (12), 14499–14509. doi:10.18632/oncotarget.7391

PubMed Abstract | CrossRef Full Text | Google Scholar

Bose, S., Barroso, M., Chheda, M. G., Clevers, H., Elez, E., Kaochar, S., et al. (2022). A path to translation: how 3D patient tumor avatars enable next generation precision oncology. Cancer Cell 40 (12), 1448–1453. doi:10.1016/j.ccell.2022.09.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Boutin, A. T., Liao, W. T., Wang, M., Hwang, S. S., Karpinets, T. V., Cheung, H., et al. (2017). Oncogenic Kras drives invasion and maintains metastases in colorectal cancer. Genes Dev. 31 (4), 370–382. doi:10.1101/gad.293449.116

PubMed Abstract | CrossRef Full Text | Google Scholar

Bray, F., Laversanne, M., Sung, H., Ferlay, J., Siegel, R. L., Soerjomataram, I., et al. (2024). Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 74 (3), 229–263. doi:10.3322/caac.21834

PubMed Abstract | CrossRef Full Text | Google Scholar

Bray, L. J., Binner, M., Holzheu, A., Friedrichs, J., Freudenberg, U., Hutmacher, D. W., et al. (2015). Multi-parametric hydrogels support 3D in vitro bioengineered microenvironment models of tumour angiogenesis. Biomaterials 53, 609–620. doi:10.1016/j.biomaterials.2015.02.124

PubMed Abstract | CrossRef Full Text | Google Scholar

Bu, P., Chen, K. Y., Xiang, K., Johnson, C., Crown, S. B., Rakhilin, N., et al. (2018). Aldolase B-mediated fructose metabolism drives metabolic reprogramming of colon cancer liver metastasis. Cell Metab. 27 (6), 1249–1262.e4. doi:10.1016/j.cmet.2018.04.003

PubMed Abstract | CrossRef Full Text | Google Scholar

Buhrmann, C., Brockmueller, A., Harsha, C., Kunnumakkara, A. B., Kubatka, P., Aggarwal, B. B., et al. (2021). Evidence that tumor microenvironment initiates epithelial-to-mesenchymal transition and calebin A can suppress it in colorectal cancer cells. Front. Pharmacol. 12, 699842. doi:10.3389/fphar.2021.699842

PubMed Abstract | CrossRef Full Text | Google Scholar

Buhrmann, C., Popper, B., Kunnumakkara, A. B., Aggarwal, B. B., and Shakibaei, M. (2019). Evidence that calebin A, a component of curcuma longa suppresses NF-B mediated proliferation, invasion and metastasis of human colorectal cancer induced by TNF-β (lymphotoxin). Nutrients 11 (12), 2904. doi:10.3390/nu11122904

PubMed Abstract | CrossRef Full Text | Google Scholar

Buzzelli, J. N., Ouaret, D., Brown, G., Allen, P. D., and Muschel, R. J. (2018). Colorectal cancer liver metastases organoids retain characteristics of original tumor and acquire chemotherapy resistance. Stem Cell Res. 27, 109–120. doi:10.1016/j.scr.2018.01.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Cañellas-Socias, A., Sancho, E., and Batlle, E. (2024). Mechanisms of metastatic colorectal cancer. Nat. Rev. Gastroenterology and Hepatology. doi:10.1038/s41575-024-00934-z

PubMed Abstract | CrossRef Full Text | Google Scholar

Cao, H., Cheng, H. S., Wang, J. K., Tan, N. S., and Tay, C. Y. (2021). A 3D physio-mimetic interpenetrating network-based platform to decode the pro and anti-tumorigenic properties of cancer-associated fibroblasts. Acta Biomater. 132, 448–460. doi:10.1016/j.actbio.2021.03.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Chandra, R., Karalis, J. D., Liu, C., Murimwa, G. Z., Voth Park, J., Heid, C. A., et al. (2021). The colorectal cancer tumor microenvironment and its impact on liver and lung metastasis. Cancers (Basel) 13 (24), 6206. doi:10.3390/cancers13246206

PubMed Abstract | CrossRef Full Text | Google Scholar

Chaudhuri, O., Koshy, S. T., Branco da Cunha, C., Shin, J.-W., Verbeke, C. S., Allison, K. H., et al. (2014). Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 13 (10), 970–978. doi:10.1038/nmat4009

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, H., Cheng, Y., Wang, X., Wang, J., Shi, X., Li, X., et al. (2020). 3D printed in vitro tumor tissue model of colorectal cancer. Theranostics 10 (26), 12127–12143. doi:10.7150/thno.52450

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, S., Boda, S. K., Batra, S. K., Li, X., and Xie, J. (2018). Emerging roles of electrospun nanofibers in cancer research. Adv. Healthc. Mater 7 (6), e1701024. doi:10.1002/adhm.201701024

PubMed Abstract | CrossRef Full Text | Google Scholar

Chen, Y., Zheng, X., and Wu, C. (2021). The role of the tumor microenvironment and treatment strategies in colorectal cancer. Front. Immunol. 12. doi:10.3389/fimmu.2021.792691

PubMed Abstract | CrossRef Full Text | Google Scholar

Coppo, R., Kondo, J., Iida, K., Okada, M., Onuma, K., Tanaka, Y., et al. (2023). Distinct but interchangeable subpopulations of colorectal cancer cells with different growth fates and drug sensitivity. iScience 26 (2), 105962. doi:10.1016/j.isci.2023.105962

PubMed Abstract | CrossRef Full Text | Google Scholar

Courau, T., Bonnereau, J., Chicoteau, J., Bottois, H., Remark, R., Assante Miranda, L., et al. (2019). Cocultures of human colorectal tumor spheroids with immune cells reveal the therapeutic potential of MICA/B and NKG2A targeting for cancer treatment. J. Immunother. Cancer 7 (1), 74. doi:10.1186/s40425-019-0553-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Crotti, S., Piccoli, M., Rizzolio, F., Giordano, A., Nitti, D., and Agostini, M. (2017). Extracellular matrix and colorectal cancer: how surrounding microenvironment affects cancer cell behavior? J. Cell Physiol. 232 (5), 967–975. doi:10.1002/jcp.25658

PubMed Abstract | CrossRef Full Text | Google Scholar

Dekker, E., Tanis, P. J., Vleugels, J. L. A., Kasi, P. M., and Wallace, M. B. (2019). Colorectal cancer. Lancet 394 (10207), 1467–1480. doi:10.1016/S0140-6736(19)32319-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Del, B. R., Pignatelli, M., Bodmer, W. F., and Wright, N. A. (1991). The role of the arginine-glycine-aspartic acid-directed cellular binding to type I collagen and rat mesenchymal cells in colorectal tumour differentiation. Differentiation 46 (2), 97–103. doi:10.1111/j.1432-0436.1991.tb00870.x

PubMed Abstract | CrossRef Full Text | Google Scholar

Deng, M., Yan, P., Gong, H., Li, G., and Wang, J. (2024). β-hydroxybutyrate resensitizes colorectal cancer cells to oxaliplatin by suppressing H3K79 methylation in vitro and in vivo. Mol. Med. 30 (1), 95. doi:10.1186/s10020-024-00864-1

PubMed Abstract | CrossRef Full Text | Google Scholar

de Visser, K. E., and Joyce, J. A. (2023). The evolving tumor microenvironment: from cancer initiation to metastatic outgrowth. Cancer Cell 41 (3), 374–403. doi:10.1016/j.ccell.2023.02.016

PubMed Abstract | CrossRef Full Text | Google Scholar

Ding, S., Hsu, C., Wang, Z., Natesh, N. R., Millen, R., Negrete, M., et al. (2022). Patient-derived micro-organospheres enable clinical precision oncology. Cell Stem Cell 29 (6), 905–917.e6. doi:10.1016/j.stem.2022.04.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Drost, J., and Clevers, H. (2018). Organoids in cancer research. Nat. Rev. Cancer 18 (7), 407–418. doi:10.1038/s41568-018-0007-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Du, J., Han, S., Zhou, H., Wang, J., Wang, F., Zhao, M., et al. (2024). Targeted protein degradation combined with PET imaging reveals the role of host PD-L1 in determining anti-PD-1 therapy efficacy. Eur. J. Nucl. Med. Mol. Imaging 51 (12), 3559–3571. doi:10.1007/s00259-024-06804-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Edmondson, R., Broglie, J. J., Adcock, A. F., and Yang, L. (2014). Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay. Drug Dev. Technol. 12 (4), 207–218. doi:10.1089/adt.2014.573

PubMed Abstract | CrossRef Full Text | Google Scholar

Fearon, E. R. (2011). Molecular genetics of colorectal cancer. Annu. Rev. Pathology Mech. Dis. 6 (6), 479–507. doi:10.1146/annurev-pathol-011110-130235

PubMed Abstract | CrossRef Full Text | Google Scholar

Fertala, A. (2020). Three decades of research on recombinant collagens: reinventing the wheel or developing new biomedical products? Bioengineering 7 (4), 155. doi:10.3390/bioengineering7040155

PubMed Abstract | CrossRef Full Text | Google Scholar

Goto, N., Westcott, P. M. K., Goto, S., Imada, S., Taylor, M. S., Eng, G., et al. (2024). SOX17 enables immune evasion of early colorectal adenomas and cancers. Nature 627 (8004), 636–645. doi:10.1038/s41586-024-07135-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Guerriero, J. L. (2018). Macrophages: the road less traveled, changing anticancer therapy. Trends Mol. Med. 24 (5), 472–489. doi:10.1016/j.molmed.2018.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

Guinney, J., Dienstmann, R., Wang, X., de Reyniès, A., Schlicker, A., Soneson, C., et al. (2015). The consensus molecular subtypes of colorectal cancer. Nat. Med. 21 (11), 1350–1356. doi:10.1038/nm.3967

PubMed Abstract | CrossRef Full Text | Google Scholar

Habanjar, O., Diab-Assaf, M., Caldefie-Chezet, F., and Delort, L. (2021). 3D cell culture systems: tumor application, advantages, and disadvantages. Int. J. Mol. Sci. 22 (22), 12200. doi:10.3390/ijms222212200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hachey, S. J., Movsesyan, S., Nguyen, Q. H., Burton-Sojo, G., Tankazyan, A., Wu, J., et al. (2021). An in vitro vascularized micro-tumor model of human colorectal cancer recapitulates in vivo responses to standard-of-care therapy. Lab. Chip 21 (7), 1333–1351. doi:10.1039/d0lc01216e

PubMed Abstract | CrossRef Full Text | Google Scholar

Hasterok, S., Gustafsson, A., and Gjörloff Wingren, A. (2023). Applications of tumor cells in an in vitro 3D environment. Appl. Sci. 13 (18), 10349. doi:10.3390/app131810349

CrossRef Full Text | Google Scholar

Health NIo (2015). NIH-wide strategic plan, fiscal years 2016–2020: turning discovery into health. Bethesda, MD: National Institutes of health (NIH), US Department of Health and Human Services[Google Scholar].

Google Scholar

Heichler, C., Scheibe, K., Schmied, A., Geppert, C. I., Schmid, B., Wirtz, S., et al. (2020). STAT3 activation through IL-6/IL-11 in cancer-associated fibroblasts promotes colorectal tumour development and correlates with poor prognosis. Gut 69 (7), 1269–1282. doi:10.1136/gutjnl-2019-319200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hirano, H., Takashima, A., Hamaguchi, T., Shida, D., and Kanemitsu, Y.Colorectal Cancer Study Group CCSG of the Japan Clinical Oncology Group JCOG (2021). Current status and perspectives of immune checkpoint inhibitors for colorectal cancer. Jpn. J. Clin. Oncol. 51 (1), 10–19. doi:10.1093/jjco/hyaa200

PubMed Abstract | CrossRef Full Text | Google Scholar

Hou, S., Zhao, Y., Chen, J., Lin, Y., and Qi, X. (2024). Tumor-associated macrophages in colorectal cancer metastasis: molecular insights and translational perspectives. J. Transl. Med. 22 (1), 62. doi:10.1186/s12967-024-04856-x

PubMed Abstract | CrossRef Full Text | Google Scholar

Ibarrola-Villava, M., Cervantes, A., and Bardelli, A. (2018). Preclinical models for precision oncology. Biochim. Biophys. Acta Rev. Cancer 1870 (2), 239–246. doi:10.1016/j.bbcan.2018.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Jacques, C., Marchesi, I., Fiorentino, F. P., Chatelais, M., Lilli, N. L., Appel, K., et al. (2022). A micro-immunotherapy sequential medicine MIM-seq displays immunomodulatory effects on human macrophages and anti-tumor properties towards in vitro 2D and 3D models of colon carcinoma and in an in vivo subcutaneous xenograft colon carcinoma model. Int. J. Mol. Sci. 23 (11), 6059. doi:10.3390/ijms23116059

PubMed Abstract | CrossRef Full Text | Google Scholar

Jensen, C., and Teng, Y. (2020). Is it time to start transitioning from 2D to 3D cell culture? Front. Mol. Biosci. 7, 33. doi:10.3389/fmolb.2020.00033

PubMed Abstract | CrossRef Full Text | Google Scholar

Jeppesen, M., Hagel, G., Glenthoj, A., Vainer, B., Ibsen, P., Harling, H., et al. (2017). Short-term spheroid culture of primary colorectal cancer cells as an in vitro model for personalizing cancer medicine. PLoS One 12 (9), e0183074. doi:10.1371/journal.pone.0183074

PubMed Abstract | CrossRef Full Text | Google Scholar

Jin, C., Wang, K., Oppong-Gyebi, A., and Hu, J. (2020). Application of nanotechnology in cancer diagnosis and therapy - a mini-review. Int. J. Med. Sci. 17 (18), 2964–2973. doi:10.7150/ijms.49801

PubMed Abstract | CrossRef Full Text | Google Scholar

Joyce, J. A., and Pollard, J. W. (2009). Microenvironmental regulation of metastasis. Nat. Rev. Cancer 9 (4), 239–252. doi:10.1038/nrc2618

PubMed Abstract | CrossRef Full Text | Google Scholar

Kai, F., Drain, A. P., and Weaver, V. M. (2019). The extracellular matrix modulates the metastatic journey. Dev. Cell 49 (3), 332–346. doi:10.1016/j.devcel.2019.03.026

PubMed Abstract | CrossRef Full Text | Google Scholar

Kawai, S., Yamazaki, M., Shibuya, K., Yamazaki, M., Fujii, E., Nakano, K., et al. (2020). Three-dimensional culture models mimic colon cancer heterogeneity induced by different microenvironments. Sci. Rep. 10 (1), 3156. doi:10.1038/s41598-020-60145-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Kawano, S., Kojima, M., Higuchi, Y., Sugimoto, M., Ikeda, K., Sakuyama, N., et al. (2015). Assessment of elasticity of colorectal cancer tissue, clinical utility, pathological and phenotypical relevance. Cancer Sci. 106 (9), 1232–1239. doi:10.1111/cas.12720

PubMed Abstract | CrossRef Full Text | Google Scholar

Kemeny, N. (2007). Presurgical chemotherapy in patients being considered for liver resection. Oncologist 12 (7), 825–839. doi:10.1634/theoncologist.12-7-825

PubMed Abstract | CrossRef Full Text | Google Scholar

Kessenbrock, K., Plaks, V., and Werb, Z. (2010). Matrix metalloproteinases: regulators of the tumor microenvironment. Cell 141 (1), 52–67. doi:10.1016/j.cell.2010.03.015

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, J., Koo, B.-K., and Knoblich, J. A. (2020). Human organoids: model systems for human biology and medicine. Nat. Rev. Mol. Cell Biol. 21 (10), 571–584. doi:10.1038/s41580-020-0259-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, M., Panagiotakopoulou, M., Chen, C., Ruiz, S. B., Ganesh, K., Tammela, T., et al. (2023). Micro-engineering and nano-engineering approaches to investigate tumour ecosystems. Nat. Rev. Cancer 23 (9), 581–599. doi:10.1038/s41568-023-00593-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Kola, I., and Landis, J. (2004). Can the pharmaceutical industry reduce attrition rates? Nat. Rev. Drug Discov. 3 (8), 711–715. doi:10.1038/nrd1470

PubMed Abstract | CrossRef Full Text | Google Scholar

Kondo, J., Ekawa, T., Endo, H., Yamazaki, K., Tanaka, N., Kukita, Y., et al. (2019). High-throughput screening in colorectal cancer tissue-originated spheroids. Cancer Sci. 110 (1), 345–355. doi:10.1111/cas.13843

PubMed Abstract | CrossRef Full Text | Google Scholar

Lawes, D. A., Pearson, T., SenGupta, S., and Boulos, P. B. (2005). The role of MLH1, MSH2 and MSH6 in the development of multiple colorectal cancers. Br. J. Cancer 93 (4), 472–477. doi:10.1038/sj.bjc.6602708

PubMed Abstract | CrossRef Full Text | Google Scholar

Lebrun, J. J. (2012). The dual role of TGFβ in human cancer: from tumor suppression to cancer metastasis. ISRN Mol. Biol. 2012, 381428. doi:10.5402/2012/381428

PubMed Abstract | CrossRef Full Text | Google Scholar

Leonard, N. A., Corry, S. M., Reidy, E., Egan, H., O'Malley, G., Thompson, K., et al. (2024). Tumor-associated mesenchymal stromal cells modulate macrophage phagocytosis in stromal-rich colorectal cancer via PD-1 signaling. iScience 27 (9), 110701. doi:10.1016/j.isci.2024.110701

PubMed Abstract | CrossRef Full Text | Google Scholar

Leonard, N. A., Reidy, E., Thompson, K., McDermott, E., Peerani, E., Tomas Bort, E., et al. (2021). Stromal cells promote matrix deposition, remodelling and an immunosuppressive tumour microenvironment in a 3D model of colon cancer. Cancers (Basel) 13 (23), 5998. doi:10.3390/cancers13235998

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C., Jiang, P., Wei, S., Xu, X., and Wang, J. (2020b). Regulatory T cells in tumor microenvironment: new mechanisms, potential therapeutic strategies and future prospects. Mol. Cancer 19 (1), 116. doi:10.1186/s12943-020-01234-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, C. Y., Wood, D. K., Hsu, C. M., and Bhatia, S. N. (2011). DNA-templated assembly of droplet-derived PEG microtissues. Lab a Chip 11 (17), 2967–2975. doi:10.1039/c1lc20318e

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, J., Liu, J., Xia, W., Yang, H., Sha, W., and Chen, H. (2024a). Deciphering the tumor microenvironment of colorectal cancer and guiding clinical treatment with patient-derived organoid technology: progress and challenges. Technol. Cancer Res. Treat. 23, 15330338231221856. doi:10.1177/15330338231221856

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, K., Liu, W., Yu, H., Chen, J., Tang, W., Wang, J., et al. (2024b). 68Ga-FAPI PET imaging monitors response to combined TGF-βR inhibition and immunotherapy in metastatic colorectal cancer. J. Clin. Invest 134 (4), e181374. doi:10.1172/JCI181374

PubMed Abstract | CrossRef Full Text | Google Scholar

Li, Z. L., Wang, Z. J., Wei, G. H., Yang, Y., and Wang, X. W. (2020a). Changes in extracellular matrix in different stages of colorectal cancer and their effects on proliferation of cancer cells. World J. Gastrointest. Oncol. 12 (3), 267–275. doi:10.4251/wjgo.v12.i3.267

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, C., Pei, H., and Tan, F. (2020). Matrix stiffness and colorectal cancer. Onco Targets Ther. 13, 2747–2755. doi:10.2147/OTT.S231010

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, D., Li, C., Deng, Z., Luo, N., Li, W., Hu, W., et al. (2024b). Multi-omics analysis reveals the landscape of tumor microenvironments in left-sided and right-sided colon cancer. Front. Med. (Lausanne) 11, 1403171. doi:10.3389/fmed.2024.1403171

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, L., Zhang, Y., Liu, H., Yang, J., Tian, Q., Chueakula, N., et al. (2025). Transforming cancer therapy: unlocking the potential of targeting vascular and stromal cells in the tumor microenvironment. Cancer Res. doi:10.1158/0008-5472.CAN-24-4744

PubMed Abstract | CrossRef Full Text | Google Scholar

Liu, Y., Wang, D., Luan, Y., Tao, B., Li, Q., Feng, Q., et al. (2024a). The application of organoids in colorectal diseases. Front. Pharmacol. 15, 1412489. doi:10.3389/fphar.2024.1412489

PubMed Abstract | CrossRef Full Text | Google Scholar

Ludwig, K., Tse, E. S., and Wang, J. Y. (2013). Colon cancer cells adopt an invasive phenotype without mesenchymal transition in 3-D but not 2-D culture upon combined stimulation with EGF and crypt growth factors. BMC Cancer 13, 221. doi:10.1186/1471-2407-13-221

PubMed Abstract | CrossRef Full Text | Google Scholar

Luo, X., Fong, E. L. S., Zhu, C., Lin, Q. X. X., Xiong, M., Li, A., et al. (2021). Hydrogel-based colorectal cancer organoid co-culture models. Acta Biomater. 132, 461–472. doi:10.1016/j.actbio.2020.12.037

PubMed Abstract | CrossRef Full Text | Google Scholar

Magdeldin, T., López-Dávila, V., Villemant, C., Cameron, G., Drake, R., Cheema, U., et al. (2014). The efficacy of cetuximab in a tissue-engineered three-dimensional in vitro model of colorectal cancer. J. Tissue Eng. 5, 2041731414544183. doi:10.1177/2041731414544183

PubMed Abstract | CrossRef Full Text | Google Scholar

Makaremi, S., Asadzadeh, Z., Hemmat, N., Baghbanzadeh, A., Sgambato, A., Ghorbaninezhad, F., et al. (2021). Immune checkpoint inhibitors in colorectal cancer: challenges and future prospects. Biomedicines 9 (9), 1075. doi:10.3390/biomedicines9091075

PubMed Abstract | CrossRef Full Text | Google Scholar

Manfredi, S., Lepage, C., Hatem, C., Coatmeur, O., Faivre, J., and Bouvier, A. M. (2006). Epidemiology and management of liver metastases from colorectal cancer. Ann. Surg. 244 (2), 254–259. doi:10.1097/01.sla.0000217629.94941.cf

PubMed Abstract | CrossRef Full Text | Google Scholar

Manfredonia, C., Muraro, M. G., Hirt, C., Mele, V., Governa, V., Papadimitropoulos, A., et al. (2019). Maintenance of primary human colorectal cancer microenvironment using a perfusion bioreactor-based 3D culture system. Adv. Biosyst. 3 (4), e1800300. doi:10.1002/adbi.201800300

PubMed Abstract | CrossRef Full Text | Google Scholar

Manou, D., Caon, I., Bouris, P., Triantaphyllidou, I. E., Giaroni, C., Passi, A., et al. (2019). The complex interplay between extracellular matrix and cells in tissues. Methods Mol. Biol. 1952, 1–20. doi:10.1007/978-1-4939-9133-4_1

PubMed Abstract | CrossRef Full Text | Google Scholar

Mao, C., Chen, Y., Xing, D., Zhang, T., Lin, Y., Long, C., et al. (2024). Resting natural killer cells promote the progress of colon cancer liver metastasis by elevating tumor-derived stem cell factor. Elife 13. doi:10.7554/eLife.97201

PubMed Abstract | CrossRef Full Text | Google Scholar

Marchalot, A., and Mjösberg, J. (2022). Innate lymphoid cells in colorectal cancer. Scand. J. Immunol. 95 (4), e13156. doi:10.1111/sji.13156

PubMed Abstract | CrossRef Full Text | Google Scholar

Marchini, A., and Gelain, F. (2022). Synthetic scaffolds for 3D cell cultures and organoids: applications in regenerative medicine. Crit. Rev. Biotechnol. 42 (3), 468–486. doi:10.1080/07388551.2021.1932716

PubMed Abstract | CrossRef Full Text | Google Scholar

Mascaux, C., Angelova, M., Vasaturo, A., Beane, J., Hijazi, K., Anthoine, G., et al. (2019). Immune evasion before tumour invasion in early lung squamous carcinogenesis. Nature 571 (7766), 570–575. doi:10.1038/s41586-019-1330-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Menyhart, O., Kakisaka, T., Pongor, L. S., Uetake, H., Goel, A., and Győrffy, B. (2019). Uncovering potential therapeutic targets in colorectal cancer by deciphering mutational status and expression of druggable oncogenes. Cancers (Basel) 11 (7), 983. doi:10.3390/cancers11070983

PubMed Abstract | CrossRef Full Text | Google Scholar

Miao, L., and Huang, L. (2015). Exploring the tumor microenvironment with nanoparticles. Cancer Treat. Res. 166, 193–226. doi:10.1007/978-3-319-16555-4_9

PubMed Abstract | CrossRef Full Text | Google Scholar

Micalet, A., Upadhyay, A., Javanmardi, Y., de Brito, C. G., Moeendarbary, E., and Cheema, U. (2024). Patient-specific colorectal-cancer-associated fibroblasts modulate tumor microenvironment mechanics. iScience 27 (6), 110060. doi:10.1016/j.isci.2024.110060

PubMed Abstract | CrossRef Full Text | Google Scholar

Mitry, E., Guiu, B., Cosconea, S., Jooste, V., Faivre, J., and Bouvier, A. M. (2010). Epidemiology, management and prognosis of colorectal cancer with lung metastases: a 30-year population-based study. Gut 59 (10), 1383–1388. doi:10.1136/gut.2010.211557

PubMed Abstract | CrossRef Full Text | Google Scholar

Miura, S., Kodaira, S., and Hosoda, Y. (1993). Immunohistologic analysis of the extracellular matrix components of the fibrous stroma of human colon cancer. J. Surg. Oncol. 53 (1), 36–42. doi:10.1002/jso.2930530111

PubMed Abstract | CrossRef Full Text | Google Scholar

Mo, S., Tang, P., Luo, W., Zhang, L., Li, Y., Hu, X., et al. (2022). Patient-derived organoids from colorectal cancer with paired liver metastasis reveal tumor heterogeneity and predict response to chemotherapy. Adv. Sci. (Weinh) 9 (31), e2204097. doi:10.1002/advs.202204097

PubMed Abstract | CrossRef Full Text | Google Scholar

Morgan, E., Arnold, M., Gini, A., Lorenzoni, V., Cabasag, C. J., Laversanne, M., et al. (2023). Global burden of colorectal cancer in 2020 and 2040: incidence and mortality estimates from GLOBOCAN. Gut 72 (2), 338–344. doi:10.1136/gutjnl-2022-327736

PubMed Abstract | CrossRef Full Text | Google Scholar

Najumudeen, A. K., Fey, S. K., Millett, L. M., Ford, C. A., Gilroy, K., Gunduz, N., et al. (2024). KRAS allelic imbalance drives tumour initiation yet suppresses metastasis in colorectal cancer in vivo. Nat. Commun. 15 (1), 100. doi:10.1038/s41467-023-44342-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Nikolaev, M., Mitrofanova, O., Broguiere, N., Geraldo, S., Dutta, D., Tabata, Y., et al. (2020). Homeostatic mini-intestines through scaffold-guided organoid morphogenesis. Nature 585 (7826), 574–578. doi:10.1038/s41586-020-2724-8

PubMed Abstract | CrossRef Full Text | Google Scholar

Nyga, A., Loizidou, M., Emberton, M., and Cheema, U. (2013). A novel tissue engineered three-dimensional in vitro colorectal cancer model. Acta Biomater. 9 (8), 7917–7926. doi:10.1016/j.actbio.2013.04.028

PubMed Abstract | CrossRef Full Text | Google Scholar

Nyström, H. (2021). Extracellular matrix proteins in metastases to the liver - composition, function and potential applications. Semin. Cancer Biol. 71, 134–142. doi:10.1016/j.semcancer.2020.06.004

PubMed Abstract | CrossRef Full Text | Google Scholar

Paduch, R., Kandefer-Szerszeń, M., and Piersiak, T. (2010). The importance of release of proinflammatory cytokines, ROS, and NO in different stages of colon carcinoma growth and metastasis after treatment with cytotoxic drugs. Oncol. Res. 18 (9), 419–436. doi:10.3727/096504010x12671222663593

PubMed Abstract | CrossRef Full Text | Google Scholar

Palucka, A. K., and Coussens Lisa, M. (2016). The basis of oncoimmunology. Cell 164 (6), 1233–1247. doi:10.1016/j.cell.2016.01.049

PubMed Abstract | CrossRef Full Text | Google Scholar

Passardi, A., Canale, M., Valgiusti, M., and Ulivi, P. (2017). Immune checkpoints as a target for colorectal cancer treatment. Int. J. Mol. Sci. 18 (6), 1324. doi:10.3390/ijms18061324

PubMed Abstract | CrossRef Full Text | Google Scholar

Paszek, M. J., Zahir, N., Johnson, K. R., Lakins, J. N., Rozenberg, G. I., Gefen, A., et al. (2005). Tensional homeostasis and the malignant phenotype. Cancer Cell 8 (3), 241–254. doi:10.1016/j.ccr.2005.08.010

PubMed Abstract | CrossRef Full Text | Google Scholar

Peela, N., Truong, D., Saini, H., Chu, H., Mashaghi, S., Ham, S. L., et al. (2017). Advanced biomaterials and microengineering technologies to recapitulate the stepwise process of cancer metastasis. Biomaterials 133, 176–207. doi:10.1016/j.biomaterials.2017.04.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Peng, Z., Lv, X., Sun, H., Zhao, L., and Huang, S. (2025). 3D tumor cultures for drug resistance and screening development in clinical applications. Mol. Cancer 24 (1), 93. doi:10.1186/s12943-025-02281-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Petrioli, R., Lorenzi, M., Aquino, A., Marsili, S., Frediani, B., Palazzuoli, V., et al. (1995). Treatment of advanced colorectal cancer with high-dose intensity folinic acid and 5-fluorouracil plus supportive care. Eur. J. Cancer 31a (12), 2105–2108. doi:10.1016/0959-8049(95)00488-2

PubMed Abstract | CrossRef Full Text | Google Scholar

Price, B. A., Marron, J. S., Mose, L. E., Perou, C. M., and Parker, J. S. (2023). Translating transcriptomic findings from cancer model systems to humans through joint dimension reduction. Commun. Biol. 6 (1), 179. doi:10.1038/s42003-023-04529-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Qu, S., Xu, R., Yi, G., Li, Z., Zhang, H., Qi, S., et al. (2024). Patient-derived organoids in human cancer: a platform for fundamental research and precision medicine. Mol. Biomed. 5 (1), 6. doi:10.1186/s43556-023-00165-9

PubMed Abstract | CrossRef Full Text | Google Scholar

Ramos, P., Carvalho, M. R., Chen, W., Yan, L. P., Zhang, C. H., He, Y. L., et al. (2023). Microphysiological systems to study colorectal cancer: state-of-the-art. Biofabrication 15 (3), 032001. doi:10.1088/1758-5090/acc279

PubMed Abstract | CrossRef Full Text | Google Scholar

Reidy, E., Leonard, N. A., Treacy, O., and Ryan, A. E. (2021). A 3D view of colorectal cancer models in predicting therapeutic responses and resistance. Cancers (Basel) 13 (2), 227. doi:10.3390/cancers13020227

PubMed Abstract | CrossRef Full Text | Google Scholar

Romero-López, M., Trinh, A. L., Sobrino, A., Hatch, M. M., Keating, M. T., Fimbres, C., et al. (2017). Recapitulating the human tumor microenvironment: colon tumor-derived extracellular matrix promotes angiogenesis and tumor cell growth. Biomaterials 116, 118–129. doi:10.1016/j.biomaterials.2016.11.034

PubMed Abstract | CrossRef Full Text | Google Scholar

Sai, B., Dai, Y., Fan, S., Wang, F., Wang, L., Li, Z., et al. (2019). Cancer-educated mesenchymal stem cells promote the survival of cancer cells at primary and distant metastatic sites via the expansion of bone marrow-derived-PMN-MDSCs. Cell Death and Dis. 10 (12), 941. doi:10.1038/s41419-019-2149-1

PubMed Abstract | CrossRef Full Text | Google Scholar

Sato, T., Vries, R. G., Snippert, H. J., van de Wetering, M., Barker, N., Stange, D. E., et al. (2009). Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature 459 (7244), 262–265. doi:10.1038/nature07935

PubMed Abstract | CrossRef Full Text | Google Scholar

Schrader, J., Gordon-Walker, T. T., Aucott, R. L., van Deemter, M., Quaas, A., Walsh, S., et al. (2011). Matrix stiffness modulates proliferation, chemotherapeutic response, and dormancy in hepatocellular carcinoma cells. Hepatology 53 (4), 1192–1205. doi:10.1002/hep.24108

PubMed Abstract | CrossRef Full Text | Google Scholar

Shah, D. D., Chorawala, M. R., Raghani, N. R., Patel, R., Fareed, M., Kashid, V. A., et al. (2025). Tumor microenvironment: recent advances in understanding and its role in modulating cancer therapies. Med. Oncol. 42 (4), 117. doi:10.1007/s12032-025-02641-4

PubMed Abstract | CrossRef Full Text | Google Scholar

Siegel, R. L., Wagle, N. S., Cercek, A., Smith, R. A., and Jemal, A. (2023). Colorectal cancer statistics, 2023. CA A Cancer J. Clin. 73 (3), 233–254. doi:10.3322/caac.21772

PubMed Abstract | CrossRef Full Text | Google Scholar

Song, K., Wang, Z., Liu, R., Chen, G., and Liu, L. (2018). Microfabrication-based three-dimensional (3-D) extracellular matrix microenvironments for cancer and other diseases. Int. J. Mol. Sci. 19 (4), 935. doi:10.3390/ijms19040935

PubMed Abstract | CrossRef Full Text | Google Scholar

Spence, J. R., Lange, A. W., Lin, S.-C. J., Kaestner, K. H., Lowy, A. M., Kim, I., et al. (2009). Sox17 regulates organ lineage segregation of ventral foregut progenitor cells. Dev. cell 17 (1), 62–74. doi:10.1016/j.devcel.2009.05.012

PubMed Abstract | CrossRef Full Text | Google Scholar

Strasser, K., Birnleitner, H., Beer, A., Pils, D., Gerner, M. C., Schmetterer, K. G., et al. (2019). Immunological differences between colorectal cancer and normal mucosa uncover a prognostically relevant immune cell profile. Oncoimmunology 8 (2), e1537693. doi:10.1080/2162402X.2018.1537693

PubMed Abstract | CrossRef Full Text | Google Scholar

Straussman, R., Morikawa, T., Shee, K., Barzily-Rokni, M., Qian, Z. R., Du, J., et al. (2012). Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487 (7408), 500–504. doi:10.1038/nature11183

PubMed Abstract | CrossRef Full Text | Google Scholar

Su, Y., Liu, J., Tian, Y., Dong, H., Shi, M., Zhang, J., et al. (2024). HIF-1α mediates immunosuppression and chemoresistance in colorectal cancer by inhibiting CXCL9, -10 and -11. Biomed. Pharmacother. 173, 116427. doi:10.1016/j.biopha.2024.116427

PubMed Abstract | CrossRef Full Text | Google Scholar

Tauriello, D. V. F., Palomo-Ponce, S., Stork, D., Berenguer-Llergo, A., Badia-Ramentol, J., Iglesias, M., et al. (2018). TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 554 (7693), 538–543. doi:10.1038/nature25492

PubMed Abstract | CrossRef Full Text | Google Scholar

Thorel, L., Perréard, M., Florent, R., Divoux, J., Coffy, S., Vincent, A., et al. (2024). Patient-derived tumor organoids: a new avenue for preclinical research and precision medicine in oncology. Exp. and Mol. Med. 56 (7), 1531–1551. doi:10.1038/s12276-024-01272-5

PubMed Abstract | CrossRef Full Text | Google Scholar

Tosca, E. M., Ronchi, D., Facciolo, D., and Magni, P. (2023). Replacement, reduction, and refinement of animal experiments in anticancer drug development: the contribution of 3D in vitro cancer models in the drug efficacy assessment. Biomedicines 11 (4), 1058. doi:10.3390/biomedicines11041058

PubMed Abstract | CrossRef Full Text | Google Scholar

Tsilimigras, D. I., Ntanasis-Stathopoulos, I., and Pawlik, T. M. (2023). Molecular mechanisms of colorectal liver metastases. Cells 12 (12), 1657. doi:10.3390/cells12121657

PubMed Abstract | CrossRef Full Text | Google Scholar

Turley, S. J., Cremasco, V., and Astarita, J. L. (2015). Immunological hallmarks of stromal cells in the tumour microenvironment. Nat. Rev. Immunol. 15 (11), 669–682. doi:10.1038/nri3902

PubMed Abstract | CrossRef Full Text | Google Scholar

Tuveson, D., and Clevers, H. (2019). Cancer modeling meets human organoid technology. Science. 364 (6444), 952–955. doi:10.1126/science.aaw6985

PubMed Abstract | CrossRef Full Text | Google Scholar

van de Wetering, M., Francies, H. E., Francis, J. M., Bounova, G., Iorio, F., Pronk, A., et al. (2015). Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell 161 (4), 933–945. doi:10.1016/j.cell.2015.03.053

PubMed Abstract | CrossRef Full Text | Google Scholar

Verma, N. K., Wong, B. H. S., Poh, Z. S., Udayakumar, A., Verma, R., Goh, R. K. J., et al. (2022). Obstacles for T-lymphocytes in the tumour microenvironment: therapeutic challenges, advances and opportunities beyond immune checkpoint. eBioMedicine 83, 104216. doi:10.1016/j.ebiom.2022.104216

PubMed Abstract | CrossRef Full Text | Google Scholar

Vitale, S., Calapà, F., Colonna, F., Luongo, F., Biffoni, M., De Maria, R., et al. (2024). Advancements in 3D in vitro models for colorectal cancer. Adv. Sci. 11 (32), 2405084. doi:10.1002/advs.202405084

PubMed Abstract | CrossRef Full Text | Google Scholar

Walker, C., Mojares, E., and Del Río Hernández, A. (2018). Role of extracellular matrix in development and cancer progression. Int. J. Mol. Sci. 19 (10), 3028. doi:10.3390/ijms19103028

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, P., Sun, L., Li, C., Jin, B., Yang, H., Wu, B., et al. (2023). Study on drug screening multicellular model for colorectal cancer constructed by three-dimensional bioprinting technology. Int. J. Bioprint 9 (3), 694. doi:10.18063/ijb.694

PubMed Abstract | CrossRef Full Text | Google Scholar

Wang, R., Mao, Y., Wang, W., Zhou, X., Wang, W., Gao, S., et al. (2022). Systematic evaluation of colorectal cancer organoid system by single-cell RNA-Seq analysis. Genome Biol. 23 (1), 106. doi:10.1186/s13059-022-02673-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Wolpin, B. M., Meyerhardt, J. A., Mamon, H. J., and Mayer, R. J. (2007). Adjuvant treatment of colorectal cancer. CA Cancer J. Clin. 57 (3), 168–185. doi:10.3322/canjclin.57.3.168

PubMed Abstract | CrossRef Full Text | Google Scholar

Xia, T., Zhang, M., Lei, W., Yang, R., Fu, S., Fan, Z., et al. (2023). Advances in the role of STAT3 in macrophage polarization. Front. Immunol. 14, 1160719. doi:10.3389/fimmu.2023.1160719

PubMed Abstract | CrossRef Full Text | Google Scholar

Yamamoto, H., Noura, S., Okami, J., Uemura, M., Takemasa, I., Ikeda, M., et al. (2008). Overexpression of MT1-MMP is insufficient to increase experimental liver metastasis of human colon cancer cells. Int. J. Mol. Med. 22 (6), 757–761.

PubMed Abstract | Google Scholar

Yan, X., Xin, Y., Yu, Y., Li, X., Li, B., Elsabahy, M., et al. (2024). Remotely controllable supramolecular nanomedicine for drug-resistant colorectal cancer therapy caused by fusobacterium nucleatum. Small Methods 8 (3), 2301309. doi:10.1002/smtd.202301309

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, H., Zhao, B., Wei, H., Zeng, H., Sheng, D., and Zhang, Y. (2022). Cucurbitacin B controls M2 macrophage polarization to suppresses metastasis via targeting JAK-2/STAT3 signalling pathway in colorectal cancer. J. Ethnopharmacol. 287, 114915. doi:10.1016/j.jep.2021.114915

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhang, X., Hu, F., Li, G., Li, G., Yang, X., Liu, L., et al. (2018). Human colorectal cancer-derived mesenchymal stem cells promote colorectal cancer progression through IL-6/JAK2/STAT3 signaling. Cell Death Dis. 9 (2), 25. doi:10.1038/s41419-017-0176-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhao, G., Liu, L., Peek, R. M., Hao, X., Polk, D. B., Li, H., et al. (2016). Activation of epidermal growth factor receptor in macrophages mediates feedback inhibition of M2 polarization and gastrointestinal tumor cell growth. J. Biol. Chem. 291 (39), 20462–20472. doi:10.1074/jbc.M116.750182

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhou, Z., Cong, L., and Cong, X. (2021). Patient-derived organoids in precision medicine: drug screening, organoid-on-a-chip and living organoid biobank. Front. Oncol. 11, 762184. doi:10.3389/fonc.2021.762184

PubMed Abstract | CrossRef Full Text | Google Scholar

Zhu, S., Niu, M., O’Mary, H., and Cui, Z. (2013). Targeting of tumor-associated macrophages made possible by PEG-sheddable, mannose-modified nanoparticles. Mol. Pharm. 10 (9), 3525–3530. doi:10.1021/mp400216r

PubMed Abstract | CrossRef Full Text | Google Scholar

Zoetemelk, M., Rausch, M., Colin, D. J., Dormond, O., and Nowak-Sliwinska, P. (2019). Short-term 3D culture systems of various complexity for treatment optimization of colorectal carcinoma. Sci. Rep. 9 (1), 7103. doi:10.1038/s41598-019-42836-0

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: colorectal cancer, in vitro, 3D model, tumor microenvironment, extracellular matrix, immune cells, therapy

Citation: Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H and Wingren AG (2025) In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front. Genet. 16:1545017. doi: 10.3389/fgene.2025.1545017

Received: 13 December 2024; Accepted: 23 April 2025;
Published: 01 May 2025.

Edited by:

Maja Sabol, Ruđer Bošković Institute, Croatia

Reviewed by:

Dahmane Oukrif, University College London, United Kingdom
Engeng Chen, Zhejiang University School of Medicine, China

Copyright © 2025 Hanitrarimalala, Prgomet, Hedhammar, Tassidis and Wingren. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Anette Gjörloff Wingren, YW5ldHRlLmdqb3Jsb2ZmLXdpbmdyZW5AbWF1LnNl

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.