Skip to main content

The progressive trend of modeling and drug screening systems of breast cancer bone metastasis

Abstract

Bone metastasis is considered as a considerable challenge for breast cancer patients. Various in vitro and in vivo models have been developed to examine this occurrence. In vitro models are employed to simulate the intricate tumor microenvironment, investigate the interplay between cells and their adjacent microenvironment, and evaluate the effectiveness of therapeutic interventions for tumors. The endeavor to replicate the latency period of bone metastasis in animal models has presented a challenge, primarily due to the necessity of primary tumor removal and the presence of multiple potential metastatic sites.

The utilization of novel bone metastasis models, including three-dimensional (3D) models, has been proposed as a promising approach to overcome the constraints associated with conventional 2D and animal models. However, existing 3D models are limited by various factors, such as irregular cellular proliferation, autofluorescence, and changes in genetic and epigenetic expression. The imperative for the advancement of future applications of 3D models lies in their standardization and automation. The utilization of artificial intelligence exhibits the capability to predict cellular behavior through the examination of substrate materials' chemical composition, geometry, and mechanical performance. The implementation of these algorithms possesses the capability to predict the progression and proliferation of cancer. This paper reviewed the mechanisms of bone metastasis following primary breast cancer. Current models of breast cancer bone metastasis, along with their challenges, as well as the future perspectives of using these models for translational drug development, were discussed.

Introduction

Breast cancer is the most common cancer worldwide, ranking first in most diagnosed malignancies in women and having the highest malignancy-related mortality [1, 2]. Despite increasing attention to screening and early detection of breast cancer, plenty of patients were only diagnosed at late stages, when multiple metastases could have been made [3, 4]. The most common sites of breast cancer metastasis include bones, lungs, liver, and brain, the most common of which is the bones and skeletal system, which account for about 65–80% of metastasis in patients with metastatic breast cancer [5, 6]. Approximately 70% of patients with advanced breast cancer developed bone metastases at the time of diagnosis [7]. Metastasis, defined by the spread and growth of malignant cells to distant organs, yields a high mortality and morbidity burden [8, 9]. Generally, when breast cancer is accompanied by metastasis, the patient’s prognosis dramatically decreases [10, 11]. Patients with metastatic breast cancer had a median overall survival of 2–3 years, with a 27% overall 5-year relative survival rate [12].

Currently, there are several treatments for bone metastases, including bone-modifying agents; however, their efficacy and side effects remain a matter of controversy [13,14,15,16]. One of the reasons for this debate could be the lack of a comprehensive understanding of the complex interplay between various involved compartments (i.e., circulating malignant cells, tumor, and bone microenvironment) [17, 18]. Therefore, understanding the mechanism through which the various stages of bone metastasis occur could play a pivotal role in the development of effective and promising treatments for advanced breast cancer.

Bone metastases are classified into two categories: osteolytic and osteoblastic, based on the interaction of malignant cells, osteoblasts, and osteoclasts. Typically, breast cancers result in osteolytic lesions; however, subsequent activation of osteoblasts could cause mixed lesions, including both the destruction of affected bone and the construction of new bone [19, 20]. Although the metastatic pattern of certain tumors depends on the characteristics of the primary cancer, there is increasing evidence of the role of the metastasis-target microenvironment in the pathophysiology of metastatic breast cancer [21]. Migrated malignant mammary cells induce the upregulation of growth factors and other cytokines in the bone microenvironment, leading to the activation of osteoclasts and inhibiting the differentiation of osteoblasts. Subsequently, pathologic bone resorption and the secretion of morphogens are responsible for the progression of the imbalance between bone formation and resorption. Moreover, chemoattractants secreted by osteocytes further direct the circulating malignant cells towards the lesions [22, 23].

As mentioned above, the development of breast cancer bone metastasis would depend on a complex interaction of various factors, which, through spatial cross-talk, eventually led to the formation of metastatic lesions in the human body [24]. Although the lack of a valid and inclusive model that could comprehensively and authentically mimic the behavior of the effector factors outside of the human body hampers the development and assessment of bone-targeted therapeutics and reduces the translational efficacy of laboratory-effective drugs [24]. Thus, in this paper, we review the mechanisms of bone metastasis following primary breast cancer. Next, we reviewed current models of breast cancer bone metastasis along with their challenges. Finally, we demonstrated the future perspectives of using these models for translational drug development.

Bone metastatic microenvironments

Bone extracellular matrix

The extracellular matrix (ECM) is a key mediator of cancer incidence, progression, and metastasis [25]. The bone extracellular matrix is a dynamic milieu composed of organic and inorganic materials. The bone ECM acts in multiple inherent (e.g., cell adhesion) or distal (e.g., calcium homeostasis) biological processes [26, 27]. The extremely high strength of bone comes from the nanocomposite structure of the bone [28]. Collagen, elastin, and polysaccharides are the major organic materials within the bone structure. Collagenous contents of various structures and molecular weights play a key role in hydroxyapatite nucleation and growth [29].

Non-collagenous proteins (about hundreds of proteins) reside in the bone ECM. They can be produced by the cells or captured from the medium by electrostatic interactions. Matrix metalloproteinases (MMP), sialoprotein, fibronectin, proteoglycans, and osteocalcin are the most important members that directly govern cellular behaviors. MMP directs matrix remodeling and impulses mesenchymal stem cells (MSCs) to osteoblasts and osteoblasts to osteocytes [30, 31]. Sialoprotein, a glycosylated and sulfated protein, interacts with cells and hydroxyapatite through RGD [32]. Proteoglycans are made from a core protein and glycosaminoglycan (GAG) family chains, which have a major role in osteogenesis. In detail, proteoglycans serve as a setting to host and release growth factors and cytokines owing to their charged characteristics [33]. Breast cancer cells expressing MMP [34], sialoproteins [35,36,37], osteocalcin [36] represent metastatic activity.

Inorganic phases of the bone possess a platelet-like morphology dispersed within the organic matrix. The mineral portion of the bone resembles the hydroxyapatite structure (Ca10(PO4)6(OH)2); however, the Ca/P ratio varies due to multiple anion (e.g., Cl) and cation (e.g., Mg2+) inclusions. Of note, the ionic substitution endows spectacular structure with individual biodegradation and bioactivity [38]. Namely, fluoride and carbonate enhance and decrease the crystallinity of bone, respectively [38]. Indeed, the constitution of the bone changes as a function of age, sex, disease, and nutrition [39].

Osteoblasts (4–6%), osteoclasts, osteocytes (90–95%) and bone lining cells are the major cells residing within the bone ECM. Mesenchymal stem cells convert into osteoclasts, osteoblasts, and bone-lining cells upon receiving a biochemical or biomechanical stimulus. In general, osteoblast proliferation/differentiation and osteoclast apoptosis/differentiation determine the bone formation ratio. Transforming growth factor-β (TGF-β), insulin-like growth factors (IGF), and fibroblast growth factors (FGF) are the most common growth factors modulating osteoblast proliferation. Meanwhile, apoptosis of the osteoclasts is mostly driven by TGF-β and drugs such as bisphosphonates and estrogen. Osteoblasts interact with ECM through integrin families such as αvβ3 (binds to RGD) / α2β1 (binds to collagen) [40, 41]. Meanwhile, osteocytes employ β1 and β3 integrins for adhering to the ECM [42, 43].

Mechanism of bone metastasis

Approximately two-thirds of patients with breast cancer are diagnosed with bone metastases. Skeletal-related events (SRE) are pathologically occurring fractures that demand surgery and cancer treatment simultaneously. Patients with > 2 cm breast cancer tumors that reside in the T and N stages were more prone to bone metastasis [44]. In a study on 295,213 patients, it was reported that hormone receptor (HR +)/human epidermal growth factor 2(HER2 +) had a high risk of bone metastasis [45]. In clinics, patients diagnosed with bone metastasis are commonly treated with bone-modifying agents (e.g., denosumab, zoledronic acid) to prevent skeletal-related events and hypercalcemia [45].

Breast cancer cells that reside in the bone ECM induce osteoclast formation by releasing tumor necrosis factor-α (TNF-α), parathyroid hormone-related peptide (PTH-rP), prostaglandin E2 (PGE2), interleukins, and leukemia inhibitory factor (LIF). PTH-rP induces osteoclast activation by receptor activator of nuclear factor-κB ligand (RANKL) cytokine secretion, which targets the RANK receptor on osteoclasts. The bone metastatic behavior of breast cancer cells was eliminated following PTH-rP neutralization [46]. Of note, RANKL is among the major inducers of osteoclast differentiation in monocytes [47] and myeloid cells. Meanwhile, RANKL expression in breast cancer cells aggravated their metastatic behavior and bone resorption via MMP1 and IL-11 activation [48, 49]. Also, cancer cells secrete TNF-α and interleukins (IL-1, IL-6, IL-8, and IL-1) which elicit osteoclast activation and migration through RANKL expression.

The bone resorption following the re-location of cancer cells brings TGF-β release, which manipulates the inhibition of osteoblast differentiation and aggravates the osteolytic activity of osteoclasts. TGF-β activates the osteolytic and metastasis-inducers PTH-rP and IL-11 [46]. PTH-rP secretion occurs following the p38 mitogen-activated protein (MAP) kinase pathway. Also, TGF- β regulates connective tissue growth factor (CTGF)/IL-11 [50], CXCR4/MMP-1 [51], and cyclooxygenase (COX)-2 [52]. Of note, TGF-β has the principal role in the epithelial-mesenchymal transition (EMT) of breast cancer cells [53].

The role of interleukins as pro-osteoclastogenesis agents is well defined by the regulation of two signaling pathways: RANK/RANKL/OPG [54] and JAK1/STAT3 signaling [55]. As lately discussed, elevated concentrations of TGF- β in bone destruction procedures increase IL production by bone marrow stromal cells, as evidenced by increased levels of IL-11 and IL-8 [56, 57]. Moreover, IL-6 serum levels vary as a function of bone metastasis and the number of additional metastasized organs [58]. The role of IL-1β [59, 60], IL-6, IL-8, and IL-11 [60] in encouraging metastatic behavior is evidenced.

Advanced modeling and drug screening systems

Solid tumors in the human body have a unique structure and behavior that enable them to escape the immune system and sometimes align the immune system with themselves to be distributed throughout the body [61]. On the other hand, they are resistant to treatment and require extensive research to discover and introduce new drugs [62].

Human body conditions' difficulties during tumor development and its interaction in laboratory simulations and the lack of a distinct tumor microenvironment (TME) model lead to limited approved anti-cancer medicine on the market [63]. So far, extensive and comprehensive efforts have been made to model tumors for drug screening and conduct more studies on tumor structure and behavior [64, 65].

Various non-tumor cells, such as endothelial cells, fibroblast cells, and immune cells, are also present in the TME and interact with tumor cells (Fig. 1). As mentioned previously, endothelial cells are essential components involved in vascular formation and tumor metastasis [66]. Fibroblasts in TME have a high proliferation rate, ECM production, and secretion of carcinogenesis-enhancing cytokines [67]. Interestingly, immune cells stimulate and inhibit the proliferation, migration, and metastasis of cancer cells [68, 69]. After the tumor escapes from the immune system, TME affects the immune cells’ function and causes the immune cells to promote the tumor spread [70]. Tumor-associated macrophages increase the ability of cancer cells to destroy the endothelial barrier [71, 72].

Fig. 1
figure 1

The tumor microenvironment consists of cancer stem cells (CSC), cancer-associated fibroblasts (CAFs), Dendritic cells (DC), myeloid-derived-suppressor cells (MDSCs), natural killer (NK), tumor-associated macrophages (TAM)

A wide range of tumor models, from monolayer 2D to 3D models and animal models, have been proposed to study cancer biology, invasiveness, metastatic disease, and drug screening [73, 74]. Currently, 2D cultures on thermoplastics and animal models are routinely used for this purpose, although each has drawbacks that limit its usage. 2D monolayer culture is an inexpensive and uncomplicated method, but it cannot reconstruct the complex 3D structure and the tumor’s multicellular components, such as immune cells [75]. This model lacks cell–cell and cell-ECM interactions as well as the cell cycle and signaling necessary for cellular functions such as proliferation, differentiation, migration, drug metabolism, and drug sensitivity [76, 77]. Due to the cells' adequate access to oxygen and nutrients, the 2D model cannot mimic the tumor’s chemical and hypoxic gradients [75].

In a native tumor, the tumor structure (hypoxia, low nutrient intake, and low pH) causes overexpression of multi drug resistance (MDR) proteins by tumor cells, resulting in the resistance of the patient's tumor cells to a wide range of chemotherapy drugs [78]. In the 2D model, it is impossible to simulate the above conditions, and the drug reaches the tumor without any physical barriers. Therefore, this model’s predictive power is not so high, and its results are not reliable for transfer to the clinic [79, 80].

Animal models also face limitations, such as time-consuming, expensive, ethical concerns, and the need to use a minimum number of animals, along with the inability to mimic specific human biology and physiology [81]. The inability of animal models to mimic the biology and physiology of the human body has limited the translation of their results to humans. Only limited manipulations can be performed on animals, and many drug screening studies have failed at the clinical phase [82]. Therefore, it is necessary to utilize PDX and immunodeficient mouse models, which are very expensive [83]. Besides, the creation of animal models is a time-consuming process and not suitable for rapid assessments.

3D models as a bridge between 2D systems and animal models can overcome the above limitations. These 3D models have evolved and have been able to mimic several features of tumor tissue, such as morphology, the gradient of chemical and biological factors, the expression of pro-angiogenic proteins, MDR, and the interactions of cell–cell and cell-ECM, via different 3D tissue engineering methods [73, 74, 84, 85]. Depending on the materials and technology used to make 3D in vitro models of bone metastasis, there are several categories. Some of the significant 3D in vitro models of bone metastasis include spheroid culture systems, scaffold-based and hydrogel-based models, bioreactor-based models, microcarrier-based models, bioprinting, and metastasis-on-a-chip systems (Table 1).

Table 1 Various invitro models of bone metastasis of breast cancer

In vitro models

Multicellular tumor spheroids

Among the scaffold-free 3D models, multicellular spheroids have become more popular so far, and there are several techniques for their construction and development [86]. These models can either be produced from a single cell-derived cancer cell line or followed by a combination of cells in specific processes, such as co-culture. The co-culture method increases the spheroids' architectural complexity and simulates the tumor structure within the body as closely as possible [63, 87].

Tumors within the body have unique characteristics, both genetically and in the cell phenotype. The mimicry of them is essential for the discovery and study of new drugs in the laboratory. Multicellular spheroids depend on cells’ location in different layers, creating a concentration gradient of nutrients, oxygen, and pH, which in turn affects cell proliferation, gene expression, and related protein translation in different layers (Fig. 1) [88]. As a result, it increases the resistance of spheroids to anti-cancer drugs. Moving from the surface to the center of this cellular sphere, the cells’ proliferative power decreases. There are also necrotic cells in the center of the cellular sphere [63, 88].

As Fig. 2a illustrates, the shape and size of spheroids varied over time. Various parameters are affecting these indexes, while time is negligible. Multicellular 3D spheroid cell cultures, possessing cytokines, vesicles, and growth factors, can develop tight junctions and ECM (Fig. 2b). The distribution of oxygen in the inner cell layers is low, so such cells have different metabolisms and metabolites than others. Accordingly, they become resistant to drugs that are sensitive to pH and oxygen changes, and these drugs lose their effectiveness (Fig. 2c). Also, the inner cell layers are usually resistant to radiation therapy due to reduced reactive oxygen species. There is a higher expression of apoptotic-resistant genes in these cells [89].

Fig. 2
figure 2

a Schematic representation of spheroid variation in shape and size over time, reprinted with permission from [88] (b) Schematic representation of a multicellular 3D spheroid cell culture, reprinted with permission from [63] (c) Main characteristics of the spheroid model, which is composed of several functionally differentiated areas and layers resulting from the impaired distribution of nutrients and oxygen. Tumor cells composing the spheroids interact with each other, developing a well-organized spatial architecture characterized by differences in phenotypic, functional, and metabolic status, reprinted with permission from [88]

Using a set of tumor cells along with stromal cells, such as fibroblasts, immune, and endothelial cells, as a collection of cells similar to the tumor microenvironment to model the spheroids creates a more heterogeneous and complex environment with cell–cell communication and signaling as well as cytokines and ECM secretions [63]. As a result, it increases tumor survival and promotes tumor cell metastasis [63].

Organotypic multicellular spheroids

In organotypic multicellular spheroids, the cells enter the spheroid condition directly based on the organ or tumor tissue culture. Thus, creating a comprehensive state of tumor modeling will preserve most of the original tumor tissue's cellular interactions. There are currently several techniques for organotypic culture; the most common is tissue slide culture [90].

Cell line culture merely for spheroid formation suffers from major obstacles, although it possesses significant advantages, such as high proliferation, inexpensiveness, and increased process speed [91]. Cell culture eventually affects gene expression and cell phenotype and causes them to deviate from their original states. Therefore, in this case, the resulting data can be somewhat unrealistic and deceptive.

This issue has been largely eliminated in organotypic culture, which provides closer information about the body's physiological environment [92]. However, organotypic culture methods are more expensive, complicated, and time-consuming than cell line culture methods. With this in mind, researchers are considering using and creating models that have both the benefits of cell and organ culture alone [63, 93]. Hence, they have started to produce new models called organoids [93].

Organoids

Organoids are essential components in creating targeted organ 3D models. Studies show that stem cells have been used to design and fabricate organoids [88, 90]. Moreover, stem cells are more stable than other differentiated cells and cell lines under ex-vivo culture conditions and can form a heterogeneous set of stem, differentiated, and functional cells. Therefore, researchers can translate the data from them for drug screening at the clinic [63, 88, 90]. Using organoids, researchers can simultaneously simulate the structure and function of healthy and tumor tissues [94,95,96]. They create a miniature of the evolutionary process of cancer in the body. In this regard, organoids can be prepared and stored from the original tumor tissues. As a result, it is possible to obtain a biobank of various tumor subtypes from different individuals (Fig. 3).

Fig. 3
figure 3

Organoids as tumor biobanks, can be prepared and stored from original tumor tissues. It is possible to obtain a biobank of various tumor subtypes from different individuals

However, the best-case study is preparing organoids from the patient's autologous tissue [97]. This method is being developed and studied under the title “personalized medicine”. Organoids are still in their infancy, and by knowing more about interstitial connections in their respective microenvironments, more structures, including blood vessels and types of inflammatory cells, can be added and engineered in these models [63, 98].

3D hydrogel/scaffold models

3D matrices based on hydrogels or porous scaffolds are another approach to creating 3D tumor models. These structures encapsulate tumor cells in the matrix material. They are capable of providing TME mechanical, structural, chemical, and physical signals to cells. In hydrogels, tumor cells can move in all directions and interact with TME. The scaffold is more rigid than hydrogels, and cells can migrate in three dimensions and interact with other cells and TME components.

Polymeric scaffolds are first synthesized and then seeded with cells, while in hydrogels, the cells can mix with the polymers before the hydrogel formation [99]. Hydrogel-based 3D models are used for various purposes, such as the study of invasion [100], migration [101], angiogenesis [102], gene expression [103, 104], tumor growth [105], and drug screening [106]. They have a porous structure that allows the release of nutrients and metabolites in the model. Additionally, hydrogels have a reasonable cost, adjustability, and the ability to provide the mechanical and biochemical support necessary for cell survival and proliferation. They are adjustable for high-throughput screening (HTS) and, as will be discussed below, are also used in 3D printing techniques and microfluidics devices.

Synthetic hydrogels have sufficient flexibility in the design of tumor ECMs [107, 108]. Polyethylene glycol (PEG)-based hydrogels are studied for cell-instructive hydrogels [109]. These hydrogel systems’ physicochemical and biological properties could be designed at the molecular level [110]. The combination of bioactive molecules in the hydrogel increases cell adhesion and exposes them to degradation by proteases secreted by cells. Therefore, they can mimic ECM-cell interactions and tissue regeneration processes. These models present an acceptable ability to study morphogenesis and tumorigenesis [106, 109,110,111].

A scaffold is a biocompatible ECM to support the attachment, growth, and morphogenesis of cells. Porous scaffolds provide the ideal environment for reconstructing the native architecture and molecular crosstalk of tumor cells. Tumor cells cultured on scaffolds have a more aggressive phenotype and are more resistant to chemotherapy [112, 113]. However, scaffold-based cultures cannot accurately identify the position of cells in the structure. Lack of proper vascular structures in the tumor model for long-term perfusion in culture, limited throughput, limited HTS, the possibility of a lack of transparency, and un-uniform distribution of cells are other drawbacks of scaffold-based culture models [73, 114,115,116].

Different natural and synthetic materials are used to make scaffold-based culture models. Kar et al. [117] used the freeze extraction method to construct scaffold-based models to evaluate breast cancer's bone metastasis. For this purpose, they used 3D scaffolds based on polycaprolactone and hydroxyapatite (HAP)/clay containing MSCs, human breast cancer cell (HBCC) lines MDA-MB-231 (MM 231), and MCF-7 cells (Fig. 4a). This 3D model provided a suitable microenvironment for cell–cell and cell–matrix interactions and maintained the metastasis potential of cells. The co-culture of MSCs and MCF-7 cells showed the formation of three-dimensional tumoroids and cancer metastasis (from mesenchymal to epithelial) (Fig. 4b-g). In addition, MDA-MB-231 cells with a high metastatic potential compared to MCF-7 cells with a low metastatic potential in these models had completely different behavior, migration potential, and invasion power, which indicates the ability of these models to evaluate the metastatic behavior of cancer cells (Fig. 4h-l).

Fig. 4
figure 4

a Schematic showing the steps of sequential culture experiment; b–d Scanning electron microscope (SEM) micrographs of sequential culture of MCF-7 cells at days (23 + 5), (23 + 10), and (23 + 15; white circles/ellipses represent tumoroids); e–g Sequential culture of MM 231 cells at days (23 + 5), (23 + 10), and (23 + 15). (Black circles/ellipses represent disorganized clusters) (X + Y days: MSCs were cultured on polycaprolactone (PCL)/in situ HAPclay scaffolds for X days, then cancer cells were seeded and culture was continued for Y more days); h–l Representative immunofluorescence microscope images of MM 231 and MCF-7 cells cultured in 2D and 3D sequential culture after immunostaining for nuclei, h E-cadherin, i vimentin, j, k vascular endothelial growth factor (VEGF), and l) cytokeratin 18. Scale bar: 50 μm. Abbreviations: Human breast cancer cell (HBCC); human breast cancer cell (HBCC) lines MDA-MB-231 (MM 231). Reprinted with permission from [117]

Moreau et al. [118] used silk scaffolds containing morphogenetic protein-2 to model breast cancer bone metastasis. They seeded the scaffolds with BMSC cells for different periods and then implanted them orthotopically in NOD/SCID mice breast cancer models (containing the patient's femur). Then, SUM1315 cells were injected into the mammary glands of mice. SUM1315 cells were observed in all scaffold-bearing BMP-2 and scaffold-bearing MSCs. However, less migration was detected in the BMP-2-MSC scaffolds, which indicated the potential of the implanted scaffolds as a site of metastasis. Jin et al. [114] used acellular breast tissue as a scaffold for culturing tumor cells. The acellular breast tissue that has been attacked by breast cancer acts as a bioactive scaffold and supports epithelial-mesenchymal transmission [114]. In general, major drawbacks such as lack of perfusion, lack of shear stress, and the high cost of large-scale production have made the mere use of hydrogel- and scaffold-based systems inefficient.

Bioreactor-based models

The bioreactor-based models, in which the culture parameters are adjustable and controllable, are very similar to in vivo conditions. Closed bioreactor systems contain precision sensors connected to controller software that control dissolved oxygen levels, temperature, pH, nutrients, and metabolic input and output currents [119, 120]. Bioreactors can generate mechanical stimuli, appropriate signals, diffusion gradients, perfusion, cell–cell, and cell-ECM interactions [121]. Continuous perfusion prevents contamination and is a time-consuming process. Bioreactors are used to make various cell suspensions and play a significant role in feeding HTS by increasing microtumor production. In this regard, various bioreactors, including static systems, stirring, rotary, hollow-fiber, and perfusion, are used [120, 121].

Stirring and static bioreactors are the most common bioreactors in tumor engineering. Stirring bioreactors create a more realistic microenvironment for tumor cell growth in three dimensions. Perfusion bioreactors have great potential in surface studies and allow them to grow under dynamic conditions using fewer media [65]. This system is suitable for high-content imaging (HCI), but HTS is arduous and needs to transfer microtumors to multi-well plates. It requires a lot of space and cost for dynamic cell culture [120, 122].

Microcarrier-based models

Furthermore, microcarrier-based models can be used to co-culture multiple cell types in a spatial arrangement to grow cells that cannot aggregate and form a 3D structure spontaneously [65]. This approach is suitable for the reconstruction of solid tumors. The porosity of microcarriers provides more surface for cell growth. It is possible to develop microcarriers from both natural and synthetic polymers. Their diameter is between 50–400 μm, and due to their surface properties, density, and chemical composition, they can enhance adhesion, proliferation, and cellular activity [123]. This approach improves drug resistance and stimulates cell–cell and cell-ECM interactions.

Small microcarriers can be combined in microfluidic devices for precise and better control of experimental conditions. High-throughput microcarrier models support the cell phenotype, and cells can produce their matrix. These models are used to evaluate cellular functions, invasion, extravasation, gene expression, immune cell response, and drug screening [124,125,126]. However, they lack perfusion, shear stress, and vasculature [123, 127, 128].

The main challenge of the above 3D in vitro models is their overly simple structure and low vascular potential. Tumor spheroids and scaffold-based tumor models have size limitations due to the lack of vessels. Although they are reliable models for tumor genesis, they are not suitable for later tumor development stages. Most of these models lack the appropriate spatial distribution of tumor cells and ECM composition. Therefore, new methods, such as bioprinting and microfluidics, have been proposed for creating realistic tumor models to improve and accelerate the diagnosis and treatment of cancer.

Bioprinting models

Bioprinting is an emerging approach to 3D modeling of cancer that makes it possible to control the temporal and spatial distribution of cells and the spatial distance between cell types [129, 130]. 3D bioprinting is a sequential (layer-by-layer) method of distributing and depositing biological components, such as bioinks and cells, in a specific position to achieve computer-designed native tumor-like structures [131]. The ECM obtained by this method can be mixed with living cells before printing or loaded with cells after printing.

Bioprinting methods have unique capabilities, such as automatic and accurate control of cell distribution in three dimensions, creating large-scale structures, high-efficiency production of cancer models, reproducibility, integration of permeable vascular networks in engineered tissues, and accurate mimicry of the complex architecture and properties of TME [132]. Using 3D bioprinting methods, various properties of TME, such as stiffness and the spatial distribution of biochemical factors, can be adjusted. The effect of the TME on different stages of development, migration, metastasis, invasion, cell–cell interactions, and cell-ECM interactions of tumor cells can be investigated.

The bioprinting technique is useful in drug screening since it mimics the morphological and genetic profiles of tumors and the non-uniform deposition of various cell types and matrix components [133, 134]. Besides, this method makes it possible to create high cell densities and in vivo interactions. Bioink containing different cell types and ECMs can print materials into large-scale tumor tissues through high-density bioprinting. The bioink used to build tumor models must be carefully selected or designed. A suitable bioink must be printable, cross-linkable, and biocompatible [131]. The stability of the structure depends on its cross-linking. Table 2 demonstrates the printed models via the bioprinting technique and the biomaterials and cells used.

Table 2 Bioprinted breast cancer models and their used biomaterials and cells

Bioinks are derived from various materials, such as natural polymers, synthetic polymers, hydrogels, decellularized ECMs (dECM), tissue spheroids, cell pellets, and microcarriers. dECM derived from patients' tissues plays a vital role in determining cell–cell and cell-ECM interactions, genetic mutations, inducing growth, and differentiation [144, 145]. The hydrogels used for bioink can create 3D architecture with physicochemical and mechanical properties that are adjustable and similar to the native tissue and preserve living cells [129, 133, 146].

The proper design of the bioink to increase cell survival is one of the most significant bioprinting challenges. Other challenges related to the bioprinting method include selecting the appropriate bioink for the tumor tissue, the final model’s dimensions, the time required for modeling, and the need to improve the resolution. The selected bioink must have both the mechanical and physiological properties necessary for the printing process and the printed tumor simultaneously. The development of specific and appropriate bioink in bioprinting can significantly improve drug screening and tumor cells’ interactions in the early stages and progression of various cancers [107, 146].

Cancer-on-a-chip systems

Cancer-on-a-chip systems are among the newest tumor models that carefully conserve the patient's tumor characteristics and can be used to select the most effective treatment for the patient. It can determine the success rate of chemotherapy drugs in less than 12 h [147]. Generally, cancer-on-a-chip platforms are 3D and multichannel microfluidic cell culture microdevices that are useful to model the physiology and biology of TME in vitro [116, 148, 149]. The integration of microfluidic technologies, microfabrication methods, and tissue engineering can help make tumor-on-chip systems.

Cancer-on-a-chip systems cause better reconstruction and precise spatio-temporal control of TME parameters. These miniature chips reduce the requirements for sample size and consumable materials during in-vitro tests and enable large-scale applications and rapid sample processing [150]. Due to the small size of the specimens, fewer animals are used for further testing, which considerably eliminates ethical concerns. It is possible to accelerate the research process by running several samples on one device.

Combining patient-derived tissues in these models allows for predicting the patient's drug response and determining the most effective drug with minimal toxicity [151]. Surgery, biopsy, aspiration, and the patient's blood sampling are proper methods for obtaining human cells [152]. A biopsy taken from a patient can spread to several tumors or spheroids that can be cultured on a tumor chip. Cancer cells and healthy cells are integrated into tumor chips and used for microclinical trials. Due to the small number of cells required and the high speed of testing on these chips, these systems provide unique tools to facilitate personal anti-cancer drug development [116, 153].

Researchers use transparent glass or polymers, such as polydimethylsiloxane (PDMS), to make these chips [154, 155]. The PDMS material used in fabricating chips is transparent, highly permeable to O2 and CO2 gases, and made of biocompatible polymers (Fig. 5a-b) [156]. PDMS allows the continuous checking of tissue constructs under a microscope to study cells' behavior and response to treatment (Fig. 5c). Soft lithography, laser cutting, and replica molding methods are conventional approaches to making chips [147, 157]. These chips mimic physiological flow, shear stress, and the delivery of nutrients and drugs like a real tumor by allowing fluid manipulation in small volumes (Fig. 5d-g) [158]. Tumor-derived cells are cultured in small chambers inside these miniature chips to mimic tumor tissue [116, 147].

Fig. 5
figure 5

Schematic illustration of the microfluidic device for multicellular tumor spheroids formation and drug screening application; a The overall schematic representation of the microfluidic chip. The bottom left displayed the schematic diagram of tumor spheroids formation on a microwell; b Top and side views of the microfluidic device; c The workflow of multicellular tumor spheroids formation and drug screening-related assay, reprinted with permission from [157]; d Schematic diagram of another microfluidic device, which consists of three tissue chambers (blue) and two microfluidic lines (red) connected to sources and sinks. Tiny communication pores (30 μm minimum diameter) allow the microfluidic lines and tissue chambers to communicate via diffusion and convection of interstitial fluid; e PDMS platform with two devices. The device on the left has been filled with colored dye to highlight the microfluidic channels. The scale bar indicates 3 mm; f The vascular tissue is created in the central chamber while the side chambers are loaded with ECM gels. The pressure and concentration gradients can be created by maintaining differential concentration or pressure in the fluidic lines; g Two days after the device is loaded with endothelial cells and fibroblasts, the endothelial cells form a network of the vessel (Green) in the central chamber. Scale bar shows 100 μm, reprinted with permission from [158]

Applications of cancer-on-a-chip platforms can be summarized as screening anti-cancer drugs and investigating the specific response of the patient's tumor to the drug [159], studying the tumorigenesis processes and the role of the TME in the progression of metastasis [160], conducting research about gene expression [161], creating an accurate mimic of TME elements, noninvasive real-time monitoring of cellular parameters, evaluation of mechanical properties of the TME [162], using it as a model of immunotherapy research [163], and ultimately personalizing cancer chemotherapy [159, 164].

As more than 90% of cancer-related deaths are due to metastasis, metastasis-on-a-chip models are also of particular importance [165]. These models can examine the invasion rate and different stages of the metastatic cascade. This model can be used to evaluate various cancer therapies, such as cell-based therapies, chemotherapy, radiation, anti-angiogenic drugs, antibodies or small molecules, electric field therapy, and targeted nanomedicine [166, 167].

Chen et al. [168] studied ductal carcinoma in situ (DCIS) formation via breast cancer modeling utilizing a cancer-on-a-chip system. They developed a biomimetic microengineering strategy to reconstitute the 3D structural organization and microenvironment of breast tumors in human cell-based in vitro models (Fig. 6). Hao et al. [167] used a bone-on-chip model to study the bone metastasis of breast cancer. This design simulated the interaction of cancer cells with the bone matrix perfectly. The unique characteristics of breast cancer colonization, previously only confirmed in vivo, were also observed in this model. This model facilitates and replicates laboratory studies of metastasis [167]. Using these chips, researchers can create various cancer-on-chip models and mimic some features in the tumor, such as vasculature, co-culture, shear stress, pressure, mechanical properties, and chemical and oxygen gradients, to better and faster predict drug responses [167].

Fig. 6
figure 6

Breast cancer-on-a-chip. a Ductal carcinoma in-situ (DCIS) formed in the lumen of the mammary duct, due to the accumulation of neoplastic epithelial cells, in the early stages of breast cancer; b Design of the DCIS microarchitecture; c Steps of creating DCIS microarchitecture: 1) separation of upper and lower cell culture chambers by collagen membrane. 2) Injection of collagen solution containing human fibroblast cells into the lower chamber 3) Continuous injection of HMF medium from the upper chamber and contraction of the gel due to the tensile force of fibroblasts 4) Create a Matrigel coating on the top surface. 5) Seeding of mammary epithelial cells. 6) Injection and adhesion of DCIS spheroids to epithelial cell, reprinted with permission from [168]

Microfluidic chips make it possible to mimic a vascular network in a tumor model. Tumor growth and metastasis depend on these networks. All the tumor nutrient and waste exchange processes, migration and metastasis of cancer cells, and delivery of drugs and immune cells to the tumor depend on the tumor’s vascular network [169]. The interaction of vasculature cells with cancer cells has a vital role in regulating TEM and cancer cell phenotype [71, 116, 170].

From the endothelial cell monolayer and circular endothelial cell tube to various cells’ perfusable networks, they can be used to mimic vascularity in cancer microfluidic chips. The monolayer of endothelial cells is efficient when the cylindrical geometry of blood vessels is not necessary. This construction process is simple, has high throughput, and can create shear stress [171]. This monolayer can be formed inside a microfluidic chip by two different methods: sprouting endothelial cells on a porous membrane and in a hydrogel. This strategy is efficacious for studying some drugs that prevent cancer cells from migrating [160, 172].

In more complex cases, two ways can happen: endothelial cells and other cells may grow on a circular scaffold and then be implanted in a microfluidic device, or endothelial cells may grow on the inner surface of a cylindrical hydrogel channel. The vascular of the first manner has mechanical properties similar to those of the natural vascular, but the vascular fabrication method is very tedious. In a second manner, the cancer cells must be cultured in hydrogel before the endothelial cell tube is made [71, 173].

According to the third approach, the sprouting of endothelial cells in the hydrogel causes an irregular vascular network similar to the capillary network. The vessels created in this mode are perfusable. These vascularized tumor chips are efficient for direct analysis of anti-angiogenic and anti-metastatic drugs, modeling the main stages of metastasis, and analyzing physiological barriers that are useful against drug delivery [174].

The lymphatic system is also involved in the spread of cancer cells. Microfluidic chips mimic this lymphatic system’s role, as well as both transluminal and luminal currents, in increasing cancer cell transmission [170]. Wang et al. [173] used sacrificial models of chitosan to make polysaccharide-cellulose-based microtubes. They implanted the resulting porous and elastic vessels in a collagen matrix. Endothelial cells were then cultured on these microtubes’ inner surface, and tumor cells were cultured in a collagen matrix. This model mimics the vascular migration of tumor cells. Nashimoto et al. [175] designed a vascularized cancer-on-a-chip platform and investigated perfusion’s effect on tumor growth and drug delivery on this platform (Fig. 7a-b). They found that perfusion in these structures could affect tumor cell culture for more than 24 h and increase their growth. The authors conducted a comprehensive examination of a tumor spheroid by utilizing confocal microscopy to observe the vascularized tumor spheroid. Figure 7c depicts a clear projection image indicating the integration of the tumor spheroid into the vascular network that originated from channels 1 and 3. Figure 7d depicts the observation of a luminal structure in the enlarged orthogonal view of the blood vessel [175].

Fig. 7
figure 7

Mimicking chemical and physical microenvironments of tumors in the microfluidic device; a In vivo, tumor vasculature serves as the route for nutrients and drugs; b Microfluidic platform to recapitulate in vivo tumor microenvironments (TMEs). Immunofluorescence images of the tumor spheroid with the vasculature; c Projection image of the tumor spheroid. Scale bar: 200 μm; d Orthogonal view from different planes (x–y, x–z, or y–z) of the confocal microscope images corresponding to the white rectangle area in c). White arrows indicate the vascular lumen. Scale bar: 50 μm. Red: RFP-HUVECs, yellow: E-cadherin (Alexa Fluor 633), blue: nuclei (Hoechst 33,342); e Drug administration to the tumor spheroids under static 56 condition. 2D projection image (average projection) reconstructed using z-stack images. Red: RFP-HUVECs, yellow: E-cadherin (MCF-7), blue: nuclei. The right column indicates high magnification views of the white rectangle areas in the left column. Scale bars: 200 μm (left column), 100 μm (right column). White arrows indicate E-cadherin-negative areas around the large blood vessels. Drug administration to the tumor spheroids under perfusion condition; f Tumor spheroids with the administration of paclitaxel (0, 5, and 50 ng/mL) at 0 h (before administration), and 48 h (after administration for 24 h and incubation with EGM-2 for 24 h). The color indicates the fluorescence of RFP-HUVECs. Scale bar: 200 μm. All the images are reprinted with permission from [175]

Also, they pointed out the impact of paclitaxel on tumor spheroids in a static environment. The tumor spheroids with vasculature are depicted in Fig. 7e subsequent to the administration of varying doses of paclitaxel. When the paclitaxel concentration was at 0 ng/mL, the spheroid maintained its spherical morphology and contained a prominent blood vessel at its core, which is analogous to the tumor spheroids observed prior to drug treatment. Conversely, the presence of an E-cadherin-negative region surrounding the sizable blood vessels within the spheroid was detected at concentrations of 5 and 50 ng/mL, as indicated by the white arrows in Fig. 7e. At a concentration of 500 ng/mL, the vascular network exhibited disconnection [175]. They reported that drug administration in the perfusion culture through the vascular network did not decrease the spheroid volume (Fig. 7f). The drug response produced by these systems’ perfusion conditions does not show a dose-dependent effect of the anti-cancer drugs relative to the static conditions [175].

Mixed co-culture microfluidics is useful for creating a heterogeneous cell growth environment in the tumor. These systems mix various cell types and culture them in one chamber. However, in this system, it is difficult to classify different types of monitored cells. In contrast, separate co-culture systems are useful for rapidly recognizing cell types cultured in the device [176]. In these systems, each cell type grows in a separate chamber, and the cells can interact with each other as the media diffuses through the culture medium between these chambers [177].

The cell–cell interactions cannot be transferred through the culture medium, which is not mimicked in this method [71, 178]. Cancer-on-a-chip systems can mimic the tumor’s mechanical properties, such as shear stress, ECM stiffness, and solid tumor elasticity. These properties play a vital role in the development and metastasis of cancer by activating mechanoreceptors. Changes in tumor tissue’s mechanical properties cause changes in the cytoskeleton, changes in the regulation of proteins, and cell behavior changes [179]. Micro- and nano-fabrication methods are efficient in creating these mechanical forces in cancer-on-a-chip systems [180].

Moreover, interstitial fluid pressure (IFP) and solid stress (SS) are two components of extracellular stress that reduce the effectiveness of cancer drugs by creating a barrier to drug delivery. Interstitial flow in the tumor causes a small shear stress, approximately 0.1 Dyn.cm−2 [181]. This shear stress has several effects on tumor development. Some of the most important of these include stimulation of oncogenic signaling pathways, upregulation of TGF-β, tightening the ECM by activating fibroblast contraction, and angiogenesis in the opposite direction of interstitial flow [182].

Microfluidic instruments generated a continuous flow of culture medium in the target cells’ direction and simulated this shear stress by using peristaltic pumps, syringe pumps, and the gravitational force due to the difference in flow height at the inlet and outlet of the microfluidic instrument. This shear stress affects the behavior of cells. Modeling this shear stress inside the tumor using microfluidic chips is of great importance [181, 183]. Studies on microfluidic systems have demonstrated the ability of these chips to simulate IFP [71].

The oxygen gradient and chemical gradient in the tumor also affect tumor growth, metastasis, and cancer treatment. An abnormal vascular network and a high density of cancer cells cause a hypoxic core in the tumor. This hypoxia reduces the toxicity of cancer drugs and is involved in malignancy progression through metastasis, so its precise reconstruction in microfluidic chips is essential for drug screening [184, 185].

Diffusion, convection, and electric fields are traditional methods of creating chemical gradients in microfluidic chips [186,187,188]. Zou et al. [189] used two channels with different inlets, one channel containing a target chemical and the other a buffer, to create chemical gradients in microfluidic chips. They seeded lung cancer cells in the chemical gradient formed in the middle of these channels. Their results showed a dynamic response of cells to chemotaxis by the Wnt signaling pathway [189]. Impermeable oxygen materials, gas supply channels near the cell chamber, and perfusing oxygen-scavenging chemicals are used to controlling the oxygen gradient in these chips. Hypoxic incubators are also useful in creating hypoxia [190, 191]. Since PDMS is highly permeable to O2 gas, another approach to controlling the oxygen gradient is to replace the PDMS with oxygen-impermeable materials [166, 191].

Acosta et al. [191] designed a microfluidic device that can mimic the tumor’s oxygen gradient and create chronic and intermittent hypoxia. They examined the migration of cancer cells on this platform and showed that hypoxia causes a more aggressive phenotype. To design this model, they used O2 gas emissions between the two gas supply channels. Aung et al. [192] developed a tumor-on-a-chip model using micro-patterning integration with microfluidics. They inserted a combination of endothelial cells and cancer spheroids into the gelatin methacrylate (GelMA) hydrogel inside the chip. They used the different motility of endothelial and cancer cells in response to a controlled morphogen gradient across the network to control the organization within microfluidic chips. They found that the migration of cancer cells depended on the location of the chemical source.

These systems are currently mostly used in research and have not yet seriously entered the clinical and industrial phases. The development of these chips in the clinical space has faced some limitations. Also, there is still no easy access to patient-derived tissues. Making these chips requires high levels of skill and experience. The integration of other techniques, such as organoid and 3D printing, into these chips is useful in their development. 3D printing technology facilitates the easier production of complex and functional chips.

In vivo models

Bone metastasis is a complex and multistep process involving a variety of signaling pathways. During metastasis, tumor cells undergo genetic and phenotypic changes and interact with other cells in the microenvironment [193, 194]. To create bone metastasis, tumor cells grow in the primary site, and by secreting various factors, they help prepare the bone for the entry of tumor cells. Next, the cells undergo an epithelial-to-mesenchymal transition. They spread in the blood circulation, and when they reach the bone, they are removed from the blood circulation and implanted in the metastatic niche of the bone. Generally, these cells undergo a period of sleep, and then, by interacting with the bone microenvironment, they help to reduce the anti-tumor immune response and create osteolytic lesions. Therefore, during metastasis, all the organisms of the body are involved [195, 196].

It is essential to provide suitable animal models to determine the pathogenesis of bone metastasis, identify suppressors and genes involved in metastasis, conduct chemotherapy studies, and provide new treatments for bone metastasis [197]. Animal models are the gold standard for studies related to metastasis and cancer treatment, but unlike humans, in whom spontaneous metastasis of breast tumors to bone is common, metastasis to bone is less common in animal models [24, 198]. In addition, ethical concerns, the high cost, and the different metabolic characteristics of animals have limited their use. To date, no model has reproduced all the genetic and phenotypic changes of breast cancer bone metastasis [199]. The type of tumor, ease of use, time, cost, extent of the immune system, and degree of similarity to the process of bone metastasis in humans are key parameters in choosing an animal model [200].

Animal models are classified based on various parameters, such as species, immune status of the host, site of implantation, and mode of tumor formation. Spontaneous tumorigenesis, carcinogenic agents, genetic manipulation, and transplantation of breast cancer cell lines have been used to produce animal models of breast cancer bone metastasis in rodents (with complete immunity and immunodeficiency) and non-rodent animals (such as Zebrafish and Drosophila melanogaster) [201]. Transplantation of tumor cells creates allograft (syngeneic) and xenograft models [202,203,204]. Xenograft tumor models, in turn, are divided into two categories: cell-derived xenografts (CDX) and patient-derived xenografts (PDX) [198, 205].

Bone metastases in breast cancer models are generally induced by injecting tumor cells into the site of metastasis (bone tissue) or other sites such as the heart [198, 206], blood circulation, adipose tissue [207], and caudal artery and veins [208, 209]. Each of these sites has a different bone metastasis efficiency. Generally, tail vein injection causes lung metastases, and intracardiac injection causes bone and brain metastases [210]. Various positive estrogen-receptor (ER +) cells (such as MCF-7) and negative estrogen-receptor (ER-) cells (such as MDA-MB-231 triple cells) have been used to model bone metastasis [211]. Generally, ER + cells use the intracardiac injection route for modeling. The creation of osteolytic and osteoblastic lesions by these cells is challenging and requires a lot of time. In contrast, ER-cells metastasize to the bone within 2–4 weeks, creating osteolytic lesions [212].

To model the growth of tumor cells in the primary site and spontaneous metastasis to bone, tumor cells can be injected into the mammary fat layers of mice, which leads to bone metastasis in 40–60% of cases [207]. The injection of breast cancer cells into the bloodstream can be used to investigate homing, dormancy, colonization, tumor growth, and interactions related to the bone microenvironment [213, 214]. Injection of human MDA-MB-231 cells or mouse E0771 cells into the left ventricle of the heart in mouse strains leads to homing in the long bones of the spine. To increase the homing of bone cells and reduce the need for intracardiac injection, MDA MB-231 subtypes obtained from repeated in vivo passages from mouse bone can also be used. Tail injection into these subcategories has led to bone metastasis in 95% of cases. MDA MB-231 subtypes gave rise to tumors in 90% of mice after injection into the tail artery, whereas MDA-IV cells gave rise to tumors in 80–90% of mice after injection into the lateral tail vein [60, 215]. These metastases had constant size and position and minimal metastasis to vital organs. Therefore, the mice maintained their health for a longer period. This method led to a reduction in the total number of animals used and related costs. It should be noted that the age of the animal (5 to 8 weeks) is also essential for achieving bone metastasis following intracardiac, intravenous, or intraarterial injection.

The intra-tibial injection is used to model the final stages of breast cancer bone metastasis and the direct interactions between tumor cells and the bone microenvironment. Injection of 10,000 4T1 cells into the tibia results in osteolytic lesions, and increased cell concentrations lead to metastasis to the femur, lungs, and forelimbs [196]. Direct injection into bone tissue accelerates modeling and ignores metastatic processes. On the other hand, routes such as the heart and blood circulation lead to the tumor cells in the bone, preparing the microenvironment for tumor development and progression. The engraftment of trabecular bone fragments taken from the patient's femoral head in mice can be used to investigate metastasis in the human bone environment. Injecting MDA-MB-231 or SUM-1315 subtypes in these mice leads to metastasis in human bone implants after four weeks of implantation [216, 217]. This method is valuable for modeling the interactions between the bone microenvironment and tumor cells in immunocompromised mice after injection. This method has the advantage of a high rate of tumor uptake in bone and is beneficial for studying genetic manipulation of the host/tumor cell environment.

However, each of these paths has limitations. Intracardiac injection of cancer cells is difficult and does not lead to specific bone metastases. On the other hand, intravenous injection generally leads to the production of lung tumors that rarely metastasize to the bones (usually metastasizing to the liver, spleen, or brain). In addition, large lung tumors mask weaker signals in other parts of the body. Injection into fat results in a low rate of metastases to bones and increases the number of animals needed. Intraosseous injections are also well controlled in terms of cell growth, but due to the need to drill bone, they cause local inflammation that does not mimic the metastatic process of cancerous bone from the circulatory system [208, 218]. Therefore, choosing your cell line and inoculation route should be based on dose–response studies before starting large animal experiments.

Based on the immune status of the host, animal models can be classified into two categories: immunocompetent and immunodeficient. Immunocompetent animals provide a complete immune system, and modeling breast cancer bone metastasis in them can evaluate the immune system's interaction with different stages of the metastatic process and anticancer agents [219]. Furthermore, these models can investigate lytic disease and mixed lesions, but these syngeneic models do not allow the use of human breast cancer cells or PDX [220]. BALB/c, C57BL/6, and FVB mice are used for mouse cell transplantation, carcinogen induction, and genetic modification [221]. Breast cancer, the bone microenvironment, and activated immune cells differ in humans and mouse. Human breast cancer metastasizes to the bone most of the time (80%), but mouse breast cancer mainly metastasizes to the lung and rarely to the bone [222]. These low rates of spontaneous bone metastasis have several reasons. Mouse and humans have biological differences. Mouse cells have more metabolic activity and a longer telomerase than human cells, which affects oncogenesis and phenotypic differences. Mouse tumors have the origin of mesenchymal tissue, and human tumors have the origin of epithelial cells. Furthermore, murine mammary tumors are not hormone-dependent, whereas most human breast tumors (especially those that metastasize to bone) are hormone-dependent and require higher concentrations of estrogen to support their growth [222, 223]. Therefore, the data from these models should be interpreted based on these differences.

To address this issue, researchers have generated bone trophic subtypes of mouse breast cancer cells through repeated passage in vivo from the bone. Injection of some of these cell lines into rodent strains resulted in successful metastasis to mouse bones (e.g., 4T1 (20%), E0771 (60–80%), and KEP (50%)) [59, 224, 225]. The injection of these cell lines into the left ventricle of the heart or bone has led to significant metastases in the bone. However, due to the rapid metastasis of syngeneic lines to the lung and its rapid growth, the primary tumors should be surgically removed to have the necessary time for metastatic detection in the bone because metastatic bone deposits are small and undetectable [24, 226].

Immunodeficient mouse models are divided based on their immunological profiles. Immunodeficient mouse models used in breast cancer bone metastasis include BALB/c nude, MF1 nude, NOD SCID, and NSG, each unique in terms of primary tumorigenesis and metastatic potential [196]. Using immunodeficient rodents helps facilitate the growth of human breast cancer cells in the host. These models help to study human cells in a host environment by eliminating confounding effects related to the animal's immune response. These mice lack a thymus and cannot produce mature T lymphocytes. Therefore, the possibility of rejecting the transplant is low for them [227]. These mice are used to study tumor cell colonization in bone, stages of metastasis, metastatic dormancy, and tumor growth [213, 228]. ER-cells with the ability to rapidly create osteolytic lesions are the first choice for the formation of bone metastasis in these models [213, 228]. However, since most types of human breast cancer that metastasize to bone are ER-positive, various studies have also used these cells. ER-positive cell metastasis to bone causes the formation of mixed lesions, and estradiol supplementation is needed to stimulate their growth in a non-human environment. These supplements change the bone microenvironment and make data interpretation difficult. ER-positive cells require times longer than 6 months (long-term dormancy in bone) to metastasize to bone in the absence of estradiol in the bone of immunodeficient mice. This model of ER-positive breast cancer contributes to our knowledge of dormancy and metastatic growth [229]. PDX xenografts also grow only in severely immunodeficient (NOD-SCID) mice. In NOD-SCID mice, the number of T and B lymphocytes, granulocytes, natural killer cells, and macrophages and their functions are reduced [198, 230]. But considering the role of the immune response in tumorigenesis and the activity of anticancer agents, these models do not mimic the body condition in the metastatic process [231, 232].

Non-rodent animals are also another option for modeling bone metastasis. Zebrafish have been developed as a non-rodent in vivo model to study human tumor growth, metastasis initiation, angiogenesis, and interaction with the microenvironment. Due to the rapid external growth of transparent zebrafish embryos and the ease of their genetic manipulation, zebrafish have become an excellent in vivo model for investigating single-cell interactions and the signaling mechanisms involved [233, 234]. Fluorescently labelled cancer cells can be transplanted into zebrafish embryos without worrying about transplant rejection. This in vivo model allows studying the behavior of various breast tumor cell lines with different bone metastatic potentials, PDX, and anti-metastatic drug treatments [234, 235]. Melanogaster is another non-rodent model for breast metastasis studies that provides a suitable platform for high-throughput genetic screening [236]. Among the above-mentioned models, PDX allows a more accurate summation of the phenotypic and genetic characteristics of tumors. Therefore, it will be discussed in detail in the next section.

Patient-derived xenograft (PDX)

The history of disease modeling for drug screening goes back to the 1950s, when researchers used different cell lines to induce disease-like conditions in animals [237]. CDX served as the gold standard model for decades; however, it has gradually become known that these models’ genetics and architecture vary significantly across different cell lines, various in vitro expansions, and various laboratory environmental conditions. Notably, the expansion of cells in vitro for months or even years causes a significant change in the cells’ molecular characteristics, and they do not resemble their parental tumors following the injection into animal bodies [238, 239].

Although the CDX has remained a valuable animal model since 1969, the PDX was established as a useful tool for mimicking human tumors [83]. Transplanting a few human tumors into small subcutaneous pockets in immunodeficient mice allows them to grow (Fig. 8) [240]. Transplanting this minced tumor of the first generation of mice into several recipient mice gives a conservative model that maintains the original human parental tumor regarding genetic, epigenetic, pathological, and molecular features (Table 3) [241, 242].

Fig. 8
figure 8

Patient-derived xenografts (PDX) and their applications

Table 3 Recent patient-derived xenografts by tumor type, immunodeficient mice, and site of implantation

Molecular and cytogenetic analysis of PDX showed significant resemblance to their parental tumors [243, 249]. The PDX response to anti-cancer treatments was highly comparable with clinical settings, which provides us with a unique opportunity to develop personalized medical treatments [250, 251]. Compared to their new comparator (organoids), in addition to their 3D propagation, the ability to evaluate them in vivo provides the conditions for researchers to study systemic changes in disease and their designated treatment effects. Also, numerous cells result from subcutaneous growth in the recipient mice's suitable environment, providing a reliable and replicable model on small and large scales [170, 252, 253].

Besides, one of the primary differences between CDX and PDX was the presence of tumor stroma, which supports the integrity and flexibility of the tumor and mediates perfusion, cell signaling, and cellular kinetics [254]. The application of PDX models is moving beyond preclinical studies. PDX models highly resemble human tumors regarding identification, monitoring, and treatment biomarkers [255]. They were incorporated into the clinical studies as avatar models of human tumors to assess the sensitivity and efficacy of anti-cancer therapies in clinical studies called co-clinical trials [256,257,258].

Currently, an increasing number of preclinical studies are using PDX models [240]. Along with the numerous advantages of PDX models, there are still a few obstacles that need to be overcome (Fig. 9). The inability to evaluate the immune system is one of them. As mentioned before, highly immunodeficient mice, such as NSG mice, are used for developing PDX models. Lack of immune system compartments, such as natural killer cells, B, and T lymphocytes in NSG mice, limits the ability to assess the effect of immune modulator drugs and immunotherapies focused on the recipient immune system, like vaccines [257].

Fig. 9
figure 9

Limitations and obstacles of patient-derived xenografts tumor models

The replacement of human tumor stroma with murine stroma is another major issue. After approximately 3–5 passages of PDX models getting ready for drug screening, the transplanted tumor stroma would be replaced wholly with murine connective tissue [259]. Due to the paracrine effects of the stroma and species-specific cytokines, this stromal replacement leads to heterogeneity in the tumor and can confound the findings [169]. Also, the tumor intake rate, which is defined as the chance of engrafting the implanted tumor pieces, was still low in different settings [260]. The use of a support matrix, including growth factors, can increase the engraftment success rate, alter the ECM regulatory interactions, and therefore artificially affect the tumor kinetics [169, 261].

Another significant parameter is time-consuming models, which need about 4–8 weeks to develop PDX models for personalized medical decision-making. This issue would limit its usage. Moreover, the site of transplantation is considered a primary factor. The surrounding environment has a significant effect on tumor behavior. Hence, implanting the tumor pieces in their original anatomic site (orthotopic) versus implantation in subcutaneous or sub-renal areas leads to different tumor behaviors, therefore altering the treatment response. Also, the implantation site plays a pivotal role in engrafting the tumors with a smaller size than the original tumors [262].

Challenges and future perspective

Bone metastasis (the most common site of breast cancer metastasis) affects the survival rate and quality of patients' lives. Therefore, providing models to understand the mechanism and mode of breast cancer's bone metastasis, drug screening, evaluation of drug release carriers, and development of new treatments to prevent the destructive effects of bone metastasis are among the main clinical challenges. So far, various in vitro and in vivo models have been developed to study breast cancer's bone metastasis.

In vitro models mimic the tumor microenvironment, investigate cell-microenvironment interactions, and evaluate tumor therapeutic responses [216]. Various 2D and 3D in vitro models have been proposed. Monolayer (2D) cultures are generally developed to study the migration and invasion of cancer cells and drug evaluations through a porous membrane [263]. In these models, the patient's cells (resulting from biopsies) can be used [264]. Animal models have also tried to model metastasis events and help in their treatment by using genetic manipulation or injection of tissues, cancer cells, and carcinogens in various animals with complete immunity and immunodeficiency. Current animal model systems generally do not represent the dormant phase of cancer cells [202, 226].

Of course, with methods such as intracardiac injection of human tumor cells in adult animals, it is possible to mimic the delay time of breast cancer's bone metastasis. However, this remains a challenge due to the need to remove the primary tumor to allow enough time for bone metastasis to develop, as well as the multiple possible sites for metastasis and their different growth kinetics [213].

Generally, immunodeficient mice are not able to mimic human immunology and stromal interactions, and expensive human mice and PDX should be used. In addition, new animal models are needed to study the bone metastasis of male breast cancers and the effect of various factors, such as menopause, on the therapeutic responses of bone metastasis. Unfortunately, these studies require more animals and more money, which is not economically and ethically justified [265].

As the next generation of bone metastasis models, 3D models (including spheroids, organoids, and scaffolds) have been proposed to overcome the problems related to 2D models (static condition) and animal models (high cost, ethical problems, and different physiology) and provide an accurate, reliable, and efficient model for evaluating bone metastasis in breast cancer. These developing models seek to provide better indicators to investigate the mechanism of metastasis and the effectiveness of drugs in vivo. Of course, current 3D models also face limitations.

Current spheroid models do not have a uniform size and, consequently, an un-uniform distribution of nutrients, leading to uneven cell growth in the spheroids [266]. The materials used in the scaffold fabrication are also effective in increasing the absorption rate and screening nature of anti-cancer drugs. In general, the current models are relatively simple and usually resemble the tumor in terms of morphology and differ from it in phenotype and heterogeneity [267]. These models are generally established with long-term culture-adapted cell lines that may not adequately match the pathology of bone metastasis [73]. Furthermore, the different culture conditions required for the various cells, the autofluorescence of the scaffold materials when imaging the cells, and the genetic and epigenetic changes of the cells over time are other problems with these systems [268]. Therefore, the standardization and automation of 3D models are requirements for their future applications. Creating more complex culture systems that can recreate organ functions and dynamic microenvironments in the future can be clinically useful for biological processes related to primary and metastatic tumors and the evaluation of their therapeutic responses to various types of drug carriers and new drugs.

Therefore, the next generation of in vitro tumor models will integrate new technologies into existing models. In fact, in future studies, systematic studies using artificial intelligence can be used to predict cell behavior based on the chemical composition, geometry, and mechanical characteristics of substrate materials [269, 270]. These algorithms provide the possibility of a quick and accurate initial description of 3D models to develop suitable substrates for the growth of cancer cells. In fact, by using these algorithms and 3D models, it is possible to predict the progress and metastasis of cancer [271].

Conclusions

The occurrence of bone metastasis poses a significant obstacle for individuals with breast cancer, and a range of in vitro and in vivo models have been established to investigate this phenomenon. In vitro models are utilized to replicate the complex tumor microenvironment, explore the interactions between cells and their surrounding microenvironment, and assess the efficacy of therapeutic interventions for tumors. The replication of the latency period of bone metastasis through animal models has posed a challenge, primarily due to the requirement of primary tumor removal and the existence of numerous potential metastatic sites. Novel bone metastasis models, such as three-dimensional (3D) models, have been suggested as a potential solution to address the limitations associated with two-dimensional (2D) models and animal models. Notwithstanding, extant 3D models are constrained by factors such as irregular cellular proliferation, autofluorescence, and alterations in genetic and epigenetic expression. Standardization and automation of 3D models are imperative for enhancing their future applications. The application of artificial intelligence has the potential to forecast cellular activity by analyzing the chemical composition, geometry, and mechanical properties of substrate materials. The utilization of these algorithms has the potential to forecast the advancement and dissemination of cancer.

Availability of data and materials

All the data that support the findings of this study are available in this manuscript.

Abbreviations

CTGF:

Connective tissue growth factor

COX:

Cyclooxygenase

CSC:

Cancer stem cell

CAFs:

Cancer associated fibroblasts

CDX:

Cell-derived xenografts

DC:

Dendritic cell

dECM:

Decellularized ECMs

DCIS:

Ductal carcinoma in-situ

EMT:

Epithelial-mesenchymal transition

ECM:

Extracellular matrix

FGF:

Fibroblast growth factors

GAG:

Glycosaminoglycan

GelMA:

Gelatin methacrylate

HTS:

High-throughput screening

HCI:

High-content imaging

IGF:

Insulin-like growth factors

IFP:

Interstitial fluid pressure

LIF:

Leukemia inhibitory factor

MDSC:

Myeloid-derived-suppressor cells

MMP:

Matrix metalloproteinases

MSC:

Mesenchymal stem cells

MAP:

Mitogen activated protein

MDR:

Multidrug resistance

NK:

Natural killer

PTH-Rp:

Parathyroid hormone-related peptide

PGE2:

Prostaglandin E2

PEG:

Polyethylene glycol

PDMS:

Polydimethylsiloxane

PDX:

Patient derived xenograft

RANKL:

Receptor activator of nuclear factor-κB ligand

SRE:

Skeletal related events

SS:

Solid stress

TGF-β:

Transforming growth factor-β

TNF-a:

Tumor necrosis factor-a

TME:

Tumor microenvironment

TAM:

Tumor associated macrophages

VEGF:

Vascular endothelial growth factor

References

  1. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, Bray F. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2021;71(3):209–49.

    Article  PubMed  Google Scholar 

  2. Roarty K, Echeverria GV. Laboratory models for investigating breast cancer therapy resistance and metastasis. Front Oncol. 2021;11:645698.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Westphal T, Gampenrieder SP, Rinnerthaler G, Greil R. Cure in metastatic breast cancer. Memo. 2018;11(3):172–9.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Scimeca M, Trivigno D, Bonfiglio R, Ciuffa S, Urbano N, Schillaci O, Bonanno E. Breast cancer metastasis to bone: From epithelial to mesenchymal transition to breast osteoblast-like cells. In: Seminars in cancer biology. 2021. Elsevier.

  5. Riggio AI, Varley KE, Welm AL. The lingering mysteries of metastatic recurrence in breast cancer. Br J Cancer. 2021;124(1):13–26.

    Article  PubMed  Google Scholar 

  6. Xiong Z, Deng G, Huang X, Li X, Xie X, Wang J, Shuang Z, Wang X. Bone metastasis pattern in initial metastatic breast cancer: a population-based study. Cancer Manag Res. 2018;10:287–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Marazzi F, Orlandi A, Manfrida S, Masiello V, Di Leone A, Massaccesi M, Moschella F, Franceschini G, Bria E, Gambacorta MA. Diagnosis and treatment of bone metastases in breast cancer: radiotherapy, local approach and systemic therapy in a guide for clinicians. Cancers. 2020;12(9):2390.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Weidle UH, Birzele F, Kollmorgen G, Ruger R. Molecular mechanisms of bone metastasis. Cancer Genom Proteom. 2016;13(1):1–12.

    CAS  Google Scholar 

  9. Rossi L, Longhitano C, Kola F, Del Grande M. State of art and advances on the treatment of bone metastases from breast cancer: a concise review. Chin Clin Oncolrnal. 2020;9(2):18.

    Article  Google Scholar 

  10. Salamanna F, Contartese D, Maglio M, Fini M. A systematic review on in vitro 3D bone metastases models: a new horizon to recapitulate the native clinical scenario? Oncotargetrnal. 2016;7(28):44803–20.

    Article  Google Scholar 

  11. Jiang ZR, Yang LH, Jin LZ, Yi LM, Bing PP, Zhou J, Yang JS. Identification of novel cuproptosis-related lncRNA signatures to predict the prognosis and immune microenvironment of breast cancer patients. Front Oncol. 2022;12:988680.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Wang B-X, Li K.-P, Yu T, Feng H-Y. Rosuvastatin promotes osteogenic differentiation of mesenchymal stem cells in the rat model of osteoporosis by the Wnt/β-catenin signal. Eur Rev Med Pharmacol Sci 2019;23(22),10161–8.

  13. Jeong H, Jeong JH, Kim JE, Ahn JH, Jung KH, Koh SJ, Cheon J, Sohn J, Kim GM, Lee KS, Sim SH, Park IH, Kim SB. Final results of the randomized phase 2 LEO trial and bone protective effects of everolimus for premenopausal hormone receptor-positive, HER2-negative metastatic breast cancer. Int J Cancerrnal. 2021;149(4):917–24.

    Article  CAS  Google Scholar 

  14. D’Oronzo S, Wood S, Brown JE. The use of bisphosphonates to treat skeletal complications in solid tumours. Bone. 2021;147:115907.

    Article  CAS  PubMed  Google Scholar 

  15. Nardin S, Mora E, Varughese FM, D’Avanzo F, Vachanaram AR, Rossi V, Saggia C, Rubinelli S, Gennari A. Breast cancer survivorship, quality of life, and late toxicities. Front Oncol. 2020;10(864):864.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Wang C-Y, Hong P-D, Wang D-H, Cherng J-H, Chang S-J, Liu C-C, Fang T-J, Wang Y-W. Polymeric gelatin scaffolds affect mesenchymal stem cell differentiation and its diverse applications in tissue engineering. Int Mol Sci. 2020;21(22):8632.

    Article  CAS  Google Scholar 

  17. Rani A, Stebbing J, Giamas G, Murphy J. Endocrine resistance in hormone receptor positive breast cancer-from mechanism to therapy. Front Endocrinol (Lausanne). 2019;10(245):245.

    Article  PubMed  Google Scholar 

  18. Fusco N, Malapelle U, Fassan M, Marchio C, Buglioni S, Zupo S, Criscitiello C, Vigneri P, Dei Tos AP, Maiorano E, Viale G. PIK3CA mutations as a molecular target for hormone receptor-positive, HER2-negative metastatic breast cancer. Front Oncol. 2021;11(562):644737.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Coleman RE, Brown J, Holen I. Bone metastases. Abeloff's Clin Oncol 2020:809–830. e3. https://doi.org/10.1016/B978-0-323-47674-4.00056-6.

  20. Ban J, Fock V, Aryee DN, Kovar H. Mechanisms, diagnosis and treatment of bone metastases. Cells. 2021;10(11):2944.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Riehl BD, Kim E, Bouzid T, Lim JY. The role of microenvironmental cues and mechanical loading milieus in breast cancer cell progression and metastasis. Front Bioeng Biotechnol. 2021;8:608526.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Ma X, Yu J. Role of the bone microenvironment in bone metastasis of malignant tumors-therapeutic implications. Cellular Oncologyrnal. 2020;43:751–61.

    Article  Google Scholar 

  23. Hofbauer LC, Bozec A, Rauner M, Jakob F, Perner S, Pantel K. Novel approaches to target the microenvironment of bone metastasis. Nat Rev Clin Oncol. 2021;18(8):488–505.

    Article  PubMed  Google Scholar 

  24. Ottewell PD, Lawson MA. Advances in murine models of breast cancer bone disease. J Cancer Metastasis Treat. 2021;7:11. https://doi.org/10.20517/2394-4722.2021.14.

  25. Cox TR. The matrix in cancer. J Nat Rev Cancer 2021;21:217–38. https://doi.org/10.1038/s41568-020-00329-7.

  26. Hussain Z, Mehmood S, Liu X, Liu Y, Wang G, Pei R. Decoding bone-inspired and cell-instructive cues of scaffolds for bone tissue engineering. J Eng Regener 2023;5(1):21–44. https://doi.org/10.1016/j.engreg.2023.10.003.

  27. Zhang J, Shen Q, Ma Y, Liu L, Jia W, Chen L, Xie J. Calcium homeostasis in Parkinson’s disease: from pathology to treatment. J Neurosci Bull. 2022;38(10):1267–70.

    Article  CAS  Google Scholar 

  28. Tertuliano OA, Greer JR. The nanocomposite nature of bone drives its strength and damage resistance. J Nature Mater. 2016;15(11):1195–202.

    Article  ADS  CAS  Google Scholar 

  29. Nudelman F, Pieterse K, George A, Bomans PH, Friedrich H, Brylka LJ, Hilbers PA, de With G, Sommerdijk NA. The role of collagen in bone apatite formation in the presence of hydroxyapatite nucleation inhibitors. J Nat Mater. 2010;9(12):1004–9.

    Article  ADS  CAS  Google Scholar 

  30. Manduca P, Castagnino A, Lombardini D, Marchisio S, Soldano S, Ulivi V, Zanotti S, Garbi C, Ferrari N, Palmieri D. Role of MT1-MMP in the osteogenic differentiation. J Bone. 2009;44(2):251–65.

    Article  CAS  Google Scholar 

  31. Buxton P, Bitar M, Gellynck K, Parkar M, Brown R, Young A, Knowles J, Nazhat S. Dense collagen matrix accelerates osteogenic differentiation and rescues the apoptotic response to MMP inhibition. J Bone. 2008;43(2):377–85.

    Article  CAS  PubMed  Google Scholar 

  32. Ganss B, Kim RH, Sodek J. Bone sialoprotein. J Crit Rev Oral Biol Med. 1999;10(1):79–98.

    Article  CAS  Google Scholar 

  33. Bi Y, Stuelten CH, Kilts T, Wadhwa S, Iozzo RV, Robey PG, Chen X-D, Young MF. Extracellular matrix proteoglycans control the fate of bone marrow stromal cells. J Biol Chem. 2005;280(34):30481–9.

    Article  CAS  PubMed  Google Scholar 

  34. Knapinska AM, Singh C, Drotleff G, Blanco D, Chai C, Schwab J, Herd A, Fields GB. Matrix metalloproteinase 13 inhibitors for modulation of osteoclastogenesis: enhancement of solubility and stability. J Chem Med Chem. 2021;16(7):1133–42.

    Article  CAS  Google Scholar 

  35. Zhang J-H, Tang J, Wang J, Ma W, Zheng W, Yoneda T, Chen J. Over-expression of bone sialoprotein enhances bone metastasis of human breast cancer cells in a mouse model. Int J Oncol. 2003;23(4):1043–8.

    CAS  PubMed  Google Scholar 

  36. Ibrahim T, Leong I, Sanchez-Sweatman O, Khokha R, Sodek J, Tenenbaum HC, Ganss B, Cheifetz S. Expression of bone sialoprotein and osteopontin in breast cancer bone metastases. J Clin Exper Metastasis. 2000;18(3):253–60.

    Article  CAS  Google Scholar 

  37. Rustamov V, Keller F, Klicks J, Hafner M, Rudolf R. Bone sialoprotein shows enhanced expression in early, high-proliferation stages of three-dimensional spheroid cell cultures of breast cancer cell line MDA-MB-231. Front Oncol. 2019;9:36.

    Article  PubMed  PubMed Central  Google Scholar 

  38. Von Euw S, Wang Y, Laurent G, Drouet C, Babonneau F, Nassif N, Azais T. Bone mineral: new insights into its chemical composition. Sci Rep. 2019;9(1):1–11.

    Google Scholar 

  39. Tang S, Zeenath U, Vashishth D. Effects of non-enzymatic glycation on cancellous bone fragility. J Bone. 2007;40(4):1144–51.

    Article  CAS  Google Scholar 

  40. Helfrich M, Nesbitt S, Lakkakorpi P, Barnes M, Bodary S, Shankar G, Mason W, Mendrick D, Väänänen H, Horton M. β1 integrins and osteoclast function: involvement in collagen recognition and bone resorption. J Bone. 1996;19(4):317–28.

    Article  CAS  Google Scholar 

  41. Helfrich MH, Nesbitt SA, Dorey EL, Horton MA. Rat osteoclasts adhere to a wide range of RGD (Arg-Gly-Asp) peptide-containing proteins, including the bone sialoproteins and fibronectin, via a β3 integrin. J Bone Mineral Res. 1992;7(3):335–43.

    Article  CAS  Google Scholar 

  42. Litzenberger JB, Kim J-B, Tummala P, Jacobs CR. β 1 integrins mediate mechanosensitive signaling pathways in osteocytes. J Calcified Tissue Int. 2010;86(4):325–32.

    Article  CAS  Google Scholar 

  43. McNamara L, Majeska R, Weinbaum S, Friedrich V, Schaffler MB. Attachment of osteocyte cell processes to the bone matrix. J Anatom Record: Adv Integr Anatom Evol Biol: Advances. 2009;292(3):355–63.

    Article  CAS  Google Scholar 

  44. Yao Y, Chu Y, Xu B, Hu Q, Song Q. Risk factors for distant metastasis of patients with primary triple-negative breast cancer. J Biosci Rep. 2019;39(6):BSR20190288. https://doi.org/10.1042/BSR20190288.

  45. Zekri J, Farag K, Yousof O, Zabani Y, Mohamed W, Ahmed GA. Bone modifying agents for patients with bone metastases from breast cancer managed in routine practice setting: treatment patterns and outcome. J Oncol Pharmacy Pract. 2020;26(4):906–11.

    Article  CAS  Google Scholar 

  46. Henderson MA, Danks JA, Slavin JL, Byrnes GB, Choong PF, Spillane JB, Hopper JL, Martin TJ. Parathyroid hormone–related protein localization in breast cancers predict improved prognosis. Cancer Res. 2006;66(4):2250–6.

    Article  CAS  PubMed  Google Scholar 

  47. Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, Tometsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, Galibert L. A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature. 1997;390(6656):175–9.

    Article  ADS  CAS  PubMed  Google Scholar 

  48. Casimiro S, Mohammad KS, Pires R, Tato-Costa J, Alho I, Teixeira R, Carvalho A, Ribeiro S, Lipton A, Guise TA. RANKL/RANK/MMP-1 molecular triad contributes to the metastatic phenotype of breast and prostate cancer cells in vitro. PloS one. 2013;8(5):e63153.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  49. Lu X, Wang Q, Hu G, Van Poznak C, Fleisher M, Reiss M, Massagué J, Kang Y. ADAMTS1 and MMP1 proteolytically engage EGF-like ligands in an osteolytic signaling cascade for bone metastasis. Genes Dev. 2009;23(16):1882–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kang Y, He W, Tulley S, Gupta GP, Serganova I, Chen C-R, Manova-Todorova K, Blasberg R, Gerald WL, Massagué J. Breast cancer bone metastasis mediated by the Smad tumor suppressor pathway. Proc Nat Acad Sci. 2005;102(39):13909–14.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  51. Dunn LK, Mohammad KS, Fournier PG, McKenna CR, Davis HW, Niewolna M, Peng XH, Chirgwin JM, Guise TA. Hypoxia and TGF-β drive breast cancer bone metastases through parallel signaling pathways in tumor cells and the bone microenvironment. PloS one. 2009;4(9):e6896.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  52. Hiraga T, Myoui A, Choi ME, Yoshikawa H, Yoneda T. Stimulation of cyclooxygenase-2 expression by bone-derived transforming growth factor-β enhances bone metastases in breast cancer. Cancer Res. 2006;66(4):2067–73.

    Article  CAS  PubMed  Google Scholar 

  53. Wardhani BW, Louisa M, Watanabe Y, Setiabudy R, Kato M. TGF-β-induced TMEPAI promotes epithelial-mesenchymal transition in doxorubicin-treated triple-negative breast cancer cells via SMAD3 and PI3K/AKT pathway alteration. Breast Cancer: Targets Ther. 2021;13:529.

    Google Scholar 

  54. Liu X-H, Kirschenbaum A, Yao S, Levine AC. Cross-talk between the interleukin-6 and prostaglandin E2 signaling systems results in enhancement of osteoclastogenesis through effects on the osteoprotegerin/receptor activator of nuclear factor-κB (RANK) LIGAND/RANK system. Endocrinology. 2005;146(4):1991–8.

    Article  CAS  PubMed  Google Scholar 

  55. Liang M, Ma Q, Ding N, Luo F, Bai Y, Kang F, Gong X, Dong R, Dai J, Dai Q. IL-11 is essential in promoting osteolysis in breast cancer bone metastasis via RANKL-independent activation of osteoclastogenesis. Cell death & disease. 2019;10(5):1–12.

    Article  Google Scholar 

  56. Kamalakar A, Bendre MS, Washam CL, Fowler TW, Carver A, Dilley JD, Bracey JW, Akel NS, Margulies AG, Skinner RA. Circulating interleukin-8 levels explain breast cancer osteolysis in mice and humans. Bone. 2014;61:176–85.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ren L, Wang X, Dong Z, Liu J, Zhang S. Bone metastasis from breast cancer involves elevated IL-11 expression and the gp130/STAT3 pathway. J Med Oncol. 2013;30(3):634.

    Article  Google Scholar 

  58. Salgado R, Junius S, Benoy I, Van Dam P, Vermeulen P, Van Marck E, Huget P, Dirix LY. Circulating interleukin-6 predicts survival in patients with metastatic breast cancer. Int J Cancer. 2003;103(5):642–6.

    Article  CAS  PubMed  Google Scholar 

  59. Tulotta C, Lefley DV, Freeman K, Gregory WM, Hanby AM, Heath PR, Nutter F, Wilkinson JM, Spicer-Hadlington AR, Liu X. Endogenous production of IL1B by breast cancer cells drives metastasis and colonization of the bone microenvironment. Clin Cancer Res. 2019;25(9):2769–82.

    Article  CAS  PubMed  Google Scholar 

  60. Nutter F, Holen I, Brown HK, Cross SS, Evans CA, Walker M, Coleman RE, Westbrook JA, Selby PJ, Brown JE. Different molecular profiles are associated with breast cancer cell homing compared with colonisation of bone: evidence using a novel bone-seeking cell line. Endocrine-related cancer. 2014;21(2):327–41.

    Article  CAS  PubMed  Google Scholar 

  61. Mansouri V, Beheshtizadeh N, Gharibshahian M, Sabouri L, Varzandeh M, Rezaei N. Recent advances in regenerative medicine strategies for cancer treatment. Biomed Pharmacother. 2021;141:111875.

    Article  CAS  PubMed  Google Scholar 

  62. Yan J, Liu D, Wang J, You W, Yang W, Yan S, He W. Rewiring chaperone-mediated autophagy in cancer by a prion-like chemical inducer of proximity to counteract adaptive immune resistance. Drug Resist Updates. 2024;73:101037.

    Article  CAS  Google Scholar 

  63. Zanoni M, Pignatta S, Arienti C, Bonafè M, Tesei A. Anticancer drug discovery using multicellular tumor spheroid models. Expert Opin Drug Discov. 2019;14(3):289–301.

    Article  CAS  PubMed  Google Scholar 

  64. Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12(4):207–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Benien P, Swami A. 3D tumor models: History, advances and future perspectives. J Fut Oncol (London, England). 2014;10:1311–27.

    Article  CAS  Google Scholar 

  66. Ameri A, Ahmed HM, Pecho RDC, Arabnozari H, Sarabadani H, Esbati R, Mirabdali S, Yazdani O. Diverse activity of miR-150 in tumor development: shedding light on the potential mechanisms. Cancer Cell Int. 2023;23(1):261.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Orimo A, Gupta PB, Sgroi DC, Arenzana-Seisdedos F, Delaunay T, Naeem R, Carey VJ, Richardson AL, Weinberg RA. Stromal fibroblasts present in invasive human breast carcinomas promote tumor growth and angiogenesis through elevated SDF-1/CXCL12 secretion. Cell. 2005;121(3):335–48.

    Article  CAS  PubMed  Google Scholar 

  68. Ma C, Fan R, Ahmad H, Shi Q, Comin-Anduix B, Chodon T, Koya RC, Liu C-C, Kwong GA, Radu CG, Ribas A, Heath JR. A clinical microchip for evaluation of single immune cells reveals high functional heterogeneity in phenotypically similar T cells. Nat Med. 2011;17(6):738–43.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Vacchelli E, Ma Y, Baracco EE, Sistigu A, Enot DP, Pietrocola F, Yang H, Adjemian S, Chaba K, Semeraro M, Signore M, De Ninno A, Lucarini V, Peschiaroli F, Businaro L, Gerardino A, Manic G, Ulas T, Günther P, Schultze JL, Kepp O, Stoll G, Lefebvre C, Mulot C, Castoldi F, Rusakiewicz S, Ladoire S, Apetoh L, Bravo-San Pedro JM, Lucattelli M, Delarasse C, Boige V, Ducreux M, Delaloge S, Borg C, André F, Schiavoni G, Vitale I, Laurent-Puig P, Mattei F, Zitvogel L, Kroemer G. Chemotherapy-induced antitumor immunity requires formyl peptide receptor 1. Science. 2015;350(6263):972–8.

    Article  CAS  PubMed  Google Scholar 

  70. Mirabdali S, Ghafouri K, Farahmand Y, Gholizadeh N, Yazdani O, Esbati R, Hajiagha BS, Rahimi A. The role and function of autophagy through signaling and pathogenetic pathways and lncRNAs in ovarian cancer. Pathol-Res Pract. 2024;253:154899.

    Article  CAS  PubMed  Google Scholar 

  71. Shang M, Soon RH, Lim CT, Khoo BL, Han J. Functionalizing the tumor microenvironment with microfluidics for anti-cancer drug development. 2019.

  72. Upadhyay S, Sharma N, Gupta KB, Dhiman M. Role of immune system in tumor progression and carcinogenesis. J Cellular Biochem. 2018;119(7):5028–42.

    Article  CAS  Google Scholar 

  73. Asghar W, El Assal R, Shafiee H, Pitteri S, Paulmurugan R, Demirci U. Engineering cancer microenvironments for in vitro 3-D tumor models. Mater Today. 2015;18(10):539–53.

    Article  CAS  Google Scholar 

  74. Stock K, Estrada MF, Vidic S, Gjerde K, Rudisch A, Santo VE, Barbier M, Blom S, Arundkar SC, Selvam I. Capturing tumor complexity in vitro: comparative analysis of 2D and 3D tumor models for drug discovery. Sci Rep. 2016;6(1):1–15.

    Article  Google Scholar 

  75. Duval K, Grover H, Han L-H, Mou Y, Pegoraro AF, Fredberg J, Chen Z. Modeling physiological events in 2D vs. 3D cell culture. J Physiol. 2017;32(4):266–77.

    Article  CAS  Google Scholar 

  76. Howes AL, Richardson RD, Finlay D, Vuori K. 3-dimensional culture Systems for Anti-Cancer Compound Profiling and High-Throughput Screening Reveal Increases in EGFR inhibitor-mediated cytotoxicity compared to monolayer culture systems. PLOS ONE. 2014;9(9):e108283.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  77. Riedl A, Schlederer M, Pudelko K, Stadler M, Walter S, Unterleuthner D, Unger C, Kramer N, Hengstschläger M, Kenner L, Pfeiffer D, Krupitza G, Dolznig H. Comparison of cancer cells in 2D vs 3D culture reveals differences in AKT–mTOR–S6K signaling and drug responses. Journal of Cell Science. 2017;130(1):203.

    CAS  PubMed  Google Scholar 

  78. Huang B, Gui M, An H, Shen J, Ye F, Ni Z, Zhan H, Che L, Lai Z, Zeng J, Peng J, Lin J. Babao dan alleviates gut immune and microbiota disorders while impacting the TLR4/MyD88/NF-кB pathway to attenuate 5-fluorouracil-induced intestinal injury. Biomed Pharmacother. 2023;166:115387.

    Article  CAS  PubMed  Google Scholar 

  79. DelNero P, Lane M, Verbridge SS, Kwee B, Kermani P, Hempstead B, Stroock A, Fischbach C. 3D culture broadly regulates tumor cell hypoxia response and angiogenesis via pro-inflammatory pathways. Biomaterials. 2015;55:110–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Faute MAd, Laurent L, Ploton D, Poupon M-F, Jardillier J-C, Bobichon H. Distinctive alterations of invasiveness, drug resistance and cell–cell organization in 3D-cultures of MCF-7, a human breast cancer cell line, and its multidrug resistant variant. J Clin Exper Metastasis. 2002;19(2):161–7.

    Article  Google Scholar 

  81. Cekanova M, Rathore K. Animal models and therapeutic molecular targets of cancer: utility and limitations. Drug Des Devel Ther. 2014;8:1911–21.

    Article  PubMed  PubMed Central  Google Scholar 

  82. Zhang B, Radisic M. Organ-on-a-chip devices advance to market. Lab Chip. 2017;17(14):2395–420.

    Article  CAS  PubMed  Google Scholar 

  83. Lai Y, Wei X, Lin S, Qin L, Cheng L, Li P. Current status and perspectives of patient-derived xenograft models in cancer research. J Hematol Oncol. 2017;10(1):106.

    Article  PubMed  PubMed Central  Google Scholar 

  84. Breslin S, O’Driscoll L. Three-dimensional cell culture: the missing link in drug discovery. Drug Discov Today. 2013;18(5–6):240–9.

    Article  CAS  PubMed  Google Scholar 

  85. Begley CG, Ellis LM. Raise standards for preclinical cancer research. Nature. 2012;483(7391):531–3.

    Article  ADS  CAS  PubMed  Google Scholar 

  86. Pinto B, Henriques AC, Silva PMA, Bousbaa H. Three-dimensional spheroids as in vitro preclinical models for cancer research. J Pharmaceut 2020;12(12):1186. https://doi.org/10.3390/pharmaceutics12121186.

  87. Weiswald L-B, Bellet D, Dangles-Marie V. Spherical cancer models in tumor biology. J Neoplasia (New York, NY). 2015;17(1):1–15.

    Article  Google Scholar 

  88. Zanoni M, Cortesi M, Zamagni A, Arienti C, Pignatta S, Tesei A. Modeling neoplastic disease with spheroids and organoids. J Hematol Oncol. 2020;13(1):97.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Shehzad A, Ravinayagam V, AlRumaih H, Aljafary M, Almohazey D, Almofty S, Al-Rashid NA, Al-Suhaimi EA. Application of three-dimensional (3D) tumor cell culture systems and mechanism of drug resistance. Curr Pharmaceut Design. 2019;25(34):3599–607.

    Article  CAS  Google Scholar 

  90. Radhakrishnan J, Varadaraj S, Dash SK, Sharma A, Verma RS. Organotypic cancer tissue models for drug screening: 3D constructs, bioprinting and microfluidic chips. Drug Discovery Today. 2020;25(5):879–90.

    Article  CAS  PubMed  Google Scholar 

  91. Katt ME, Placone AL, Wong AD, Xu ZS, Searson PC. In vitro tumor models: advantages, disadvantages, variables, and selecting the right platform. Front Bioeng Biotechnol. 2016;4:12.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nature Rev Mol Cell Biol. 2014;15(10):647–64.

    Article  CAS  Google Scholar 

  93. Nishida-Aoki N, Bondesson A, Gujral T. Measuring real-time drug response in organotypic tumor tissue slices. J Visualized Exper. 2020;159. https://doi.org/10.3791/61036.

  94. Kim J, Koo B-K, Knoblich JA. Human organoids: model systems for human biology and medicine. Nat Rev Mol Cell Biol. 2020;21(10):571–84.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Simian M, Bissell MJ. Organoids: a historical perspective of thinking in three dimensions. J Cell Biol. 2016;216(1):31–40.

    Article  PubMed  Google Scholar 

  96. Kretzschmar K. Cancer research using organoid technology. J Mol Med. 2021; 99:501–15. https://doi.org/10.1007/s00109-020-01990-z.

  97. Fujii M, Sato T. Somatic cell-derived organoids as prototypes of human epithelial tissues and diseases. Nature Mater. 2021;20:156–69. https://doi.org/10.1038/s41563-020-0754-0.

  98. Wörsdörfer P, Asahina TII, Sumita Y, Ergün S. Do not keep it simple: recent advances in the generation of complex organoids. J Neur Transmis. 2020;127(11):1569–77.

    Article  Google Scholar 

  99. Guven S, Chen P, Inci F, Tasoglu S, Erkmen B, Demirci U. Multiscale assembly for tissue engineering and regenerative medicine. Trends Biotechnol. 2015;33(5):269–79.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Liu C, Lewin Mejia D, Chiang B, Luker KE, Luker GD. Hybrid collagen alginate hydrogel as a platform for 3D tumor spheroid invasion. J Acta Biomater. 2018;75:213–25.

    Article  ADS  CAS  Google Scholar 

  101. Huang Y, Tong L, Yi L, Zhang C, Hai L, Li T, Yu S, Wang W, Tao Z, Ma H, Liu P, Xie Y, Yang X. Three-dimensional hydrogel is suitable for targeted investigation of amoeboid migration of glioma cells. Mol Med Rep. 2018;17(1):250–6.

    CAS  PubMed  Google Scholar 

  102. Song HH, Park KM, Gerecht S. Hydrogels to model 3D in vitro microenvironment of tumor vascularization. Adv Drug Deliv Rev. 2014;79–80:19–29.

    Article  PubMed  Google Scholar 

  103. Ngo MT, Harley BAC. Perivascular signals alter global gene expression profile of glioblastoma and response to temozolomide in a gelatin hydrogel. Biomaterials. 2019;198:122–34. https://doi.org/10.1016/j.biomaterials.2018.06.013.

  104. Tang L, Li J, Bao M, Xiang J, Chen Y, Wang Y. Genetic association between HER2 and ESR2 polymorphisms and ovarian cancer: a meta-analysis. Onco Targets Ther. 2018;11:1055–66.

    Article  PubMed  PubMed Central  Google Scholar 

  105. Li Y, Kumacheva E. Hydrogel microenvironments for cancer spheroid growth and drug screening. JouSci Adv. 2018;4(4):eaas8998.

    ADS  Google Scholar 

  106. Fong EL, Martinez M, Yang J, Mikos AG, Navone NM, Harrington DA, Farach-Carson MC. Hydrogel-based 3D model of patient-derived prostate xenograft tumors suitable for drug screening. Mol Pharmaceut. 2014;11(7):2040–50.

    Article  CAS  Google Scholar 

  107. Knowlton S, Onal S, Yu CH, Zhao JJ, Tasoglu S. Bioprinting for cancer research. Trends Biotechnol. 2015;33(9):504–13.

    Article  CAS  PubMed  Google Scholar 

  108. Liaw CY, Ji S, Guvendiren M. Engineering 3D hydrogels for personalized in vitro human tissue models. Adv Healthcare Mater. 2018;7(4):1701165.

    Article  Google Scholar 

  109. Liang Y, Jeong J, DeVolder RJ, Cha C, Wang F, Tong YW, Kong H. A cell-instructive hydrogel to regulate malignancy of 3D tumor spheroids with matrix rigidity. Biomaterials. 2011;32(35):9308–15.

    Article  CAS  PubMed  Google Scholar 

  110. Loessner D, Stok KS, Lutolf MP, Hutmacher DW, Clements JA, Rizzi SC. Bioengineered 3D platform to explore cell–ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials. 2010;31(32):8494–506.

    Article  CAS  PubMed  Google Scholar 

  111. Hutmacher DW. Biomaterials offer cancer research the third dimension. Nat Mater. 2010;9(2):90–3.

    Article  ADS  CAS  PubMed  Google Scholar 

  112. Fischbach C, Chen R, Matsumoto T, Schmelzle T, Brugge JS, Polverini PJ, Mooney DJ. Engineering tumors with 3D scaffolds. Nature Methods. 2007;4(10):855–60.

    Article  CAS  PubMed  Google Scholar 

  113. Lü WD, Zhang L, Wu CL, Liu ZG, Lei GY, Liu J, Gao W, Hu YR. Development of an acellular tumor extracellular matrix as a three-dimensional scaffold for tumor engineering. PLoS One. 2014;9(7):e103672.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  114. Jin Q, Liu G, Li S, Yuan H, Yun Z, Zhang W, Zhang S, Dai Y, Ma Y. Decellularized breast matrix as bioactive microenvironment for in vitro three-dimensional cancer culture. J Cell Physiol. 2019;234(4):3425–35.

    Article  CAS  PubMed  Google Scholar 

  115. Pradhan S, Hassani I, Clary JM, Lipke EA. Polymeric biomaterials for in vitro cancer tissue engineering and drug testing applications. Tissue Eng Part B: Rev. 2016;22(6):470–84.

    Article  CAS  PubMed  Google Scholar 

  116. Tsai H-F, Trubelja A, Shen AQ, Bao G. Tumour-on-a-chip: microfluidic models of tumour morphology, growth and microenvironment. J Royal Soc Interface. 2017;14(131):20170137.

    Article  Google Scholar 

  117. Kar S, Molla MS, Katti DR, Katti KS. Tissue-engineered nanoclay-based 3D in vitro breast cancer model for studying breast cancer metastasis to bone. J Tissue Eng Regener Med. 2019;13(2):119–30.

    Article  CAS  Google Scholar 

  118. Moreau JE, Anderson K, Mauney JR, Nguyen T, Kaplan DL, Rosenblatt M. Tissue-engineered bone serves as a target for metastasis of human breast cancer in a mouse model. Cancer Res. 2007;67(21):10304–8.

    Article  CAS  PubMed  Google Scholar 

  119. Cacopardo L, Costa J, Giusti S, Buoncompagni L, Meucci S, Corti A, Mattei G, Ahluwalia A. Real-time cellular impedance monitoring and imaging of biological barriers in a dual-flow membrane bioreactor. Biosens Bioelectr. 2019;140:111340.

    Article  CAS  Google Scholar 

  120. Guller A, Grebenyuk P, Shekhter A, Zvyagin A, Deyev S. Bioreactor-based tumor tissue engineering. J Acta Naturae (aнглoязычнaя вepcия). 2016;8(3):30.

    Google Scholar 

  121. Hickman JA, Graeser R, de Hoogt R, Vidic S, Brito C, Gutekunst M, van der Kuip H. Three-dimensional models of cancer for pharmacology and cancer cell biology: capturing tumor complexity in vitro/ex vivo. Biotechnol J. 2014;9(9):1115–28.

    Article  CAS  PubMed  Google Scholar 

  122. Santo VE, Estrada MF, Rebelo SP, Abreu S, Silva I, Pinto C, Veloso SC, Serra AT, Boghaert E, Alves PM. Adaptable stirred-tank culture strategies for large scale production of multicellular spheroid-based tumor cell models. J Biotechnol. 2016;221:118–29.

    Article  CAS  PubMed  Google Scholar 

  123. Sun L-Y, Lin S-Z, Li Y-S, Harn H-J, Chiou T-W. Functional cells cultured on microcarriers for use in regenerative medicine research. Cell Transplantation. 2011;20(1):49–62.

    Article  ADS  CAS  PubMed  Google Scholar 

  124. Maurer BJ, Ihnat MA, Morgan C, Pullman J, O’Brien C, Johnson SW, Rasey JS, Cornwell MM. Growth of human tumor cells in macroporous microcarriers results in p53-independent, decreased cisplatin sensitivity relative to monolayers. Mol Pharmacol. 1999;55(5):938–47.

    CAS  PubMed  Google Scholar 

  125. Brancato V, Gioiella F, Profeta M, Imparato G, Guarnieri D, Urciuolo F, Melone P, Netti PA. 3D tumor microtissues as an in vitro testing platform for microenvironmentally-triggered drug delivery systems. Acta Biomater. 2017;57:47–58.

    Article  CAS  PubMed  Google Scholar 

  126. Wu Y, Sun W, Kong Y, Liu B, Zeng M, Wang W. Restoration of microRNA-130b expression suppresses osteosarcoma cell malignant behavior in vitro. Oncol Lett. 2018;16(1):97–104.

    PubMed  PubMed Central  Google Scholar 

  127. Gioiella F, Urciuolo F, Imparato G, Brancato V, Netti PA. An engineered breast cancer model on a chip to replicate ECM-activation in vitro during tumor progression. Adv Healthcare Mater. 2016;5(23):3074–84.

    Article  CAS  Google Scholar 

  128. Brancato V, Comunanza V, Imparato G, Corà D, Urciuolo F, Noghero A, Bussolino F, Netti PA. Bioengineered tumoral microtissues recapitulate desmoplastic reaction of pancreatic cancer. Acta Biomaterialia. 2017;49:152–66.

    Article  CAS  PubMed  Google Scholar 

  129. Zhao Y, Yao R, Ouyang L, Ding H, Zhang T, Zhang K, Cheng S, Sun W. Three-dimensional printing of Hela cells for cervical tumor model in vitro. Biofabrication. 2014;6(3):035001.

    Article  ADS  CAS  PubMed  Google Scholar 

  130. Ceyhan E, Xu F, Gurkan UA, Emre AE, Turali ES, El Assal R, Acikgenc A, Wu CaM, Demirci U. Prediction and control of number of cells in microdroplets by stochastic modeling. Lab Chip. 2012;12(22):4884–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Beheshtizadeh N, Lotfibakhshaiesh N, Pazhouhnia Z, Hoseinpour M, Nafari M. A review of 3D bio-printing for bone and skin tissue engineering: a commercial approach. Jou of Materials Science. 2020;55(9):3729–49.

    Article  ADS  CAS  Google Scholar 

  132. Beheshtizadeh N, Azami M, Abbasi H, Farzin A. Applying extrusion-based 3D printing technique accelerates fabricating complex biphasic calcium phosphate-based scaffolds for bone tissue regeneration. J Advanced Res. 2022;40:69–94.

    Article  CAS  Google Scholar 

  133. Hermida MA, Kumar JD, Schwarz D, Laverty KG, Di Bartolo A, Ardron M, Bogomolnijs M, Clavreul A, Brennan PM, Wiegand UK, Melchels FPW, Shu W, Leslie NR. Three dimensional in vitro models of cancer: Bioprinting multilineage glioblastoma models. Adv Biol Regul. 2020;75:100658.

    Article  CAS  PubMed  Google Scholar 

  134. Mollica PA, Booth-Creech EN, Reid JA, Zamponi M, Sullivan SM, Palmer X-L, Sachs PC, Bruno RD. 3D bioprinted mammary organoids and tumoroids in human mammary derived ECM hydrogels. Acta Biomaterialia. 2019;95:201–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Wang Y, Shi W, Kuss M, Mirza S, Qi D, Krasnoslobodtsev A, Zeng J, Band H, Band V, Duan B. 3D bioprinting of breast cancer models for drug resistance study. ACS Biomater Sci Eng. 2018;4(12):4401–11.

    Article  CAS  PubMed  Google Scholar 

  136. Ling K, Huang G, Liu J, Zhang X, Ma Y, Lu T, Xu F. Bioprinting-based high-throughput fabrication of three-dimensional MCF-7 human breast cancer cellular spheroids. Engineering. 2015;1(2):269–74.

    Article  CAS  Google Scholar 

  137. Zhou X, Zhu W, Nowicki M, Miao S, Cui H, Holmes B, Glazer RI, Zhang LG. 3D bioprinting a cell-laden bone matrix for breast cancer metastasis study. ACS Appl Mater Interfaces. 2016;8(44):30017–26.

    Article  CAS  PubMed  Google Scholar 

  138. Zhu W, Holmes B, Glazer RI, Zhang LG. 3D printed nanocomposite matrix for the study of breast cancer bone metastasis. J Nanomed: Nanotechnol Biol Med. 2016;12(1):69–79.

    Article  CAS  Google Scholar 

  139. Zhu W, Castro NJ, Cui H, Zhou X, Boualam B, McGrane R, Glazer RI, Zhang LG. A 3D printed nano bone matrix for characterization of breast cancer cell and osteoblast interactions. Nanotechnology. 2016;27(31):315103.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  140. Lee C, Abelseth E, De La Vega L, Willerth S. Bioprinting a novel glioblastoma tumor model using a fibrin-based bioink for drug screening. Mater Today Chem. 2019;12:78–84.

    Article  CAS  Google Scholar 

  141. Cui H, Esworthy T, Zhou X, Hann SY, Glazer RI, Li R, Zhang LG. Engineering a novel 3D printed vascularized tissue model for investigating breast cancer metastasis to bone. Adv Healthcare Mater. 2020;9(15):1900924.

    Article  CAS  Google Scholar 

  142. Holmes B, Zhu W, Zhang LG. Development of a novel 3D bioprinted in vitro nano bone model for breast cancer bone metastasis study. MRS Online Proc Library (OPL). 2014;1724:mrsf14-1724-h09-03.

    Article  Google Scholar 

  143. Wang X, Li X, Dai X, Zhang X, Zhang J, Xu T, Lan Q. Coaxial extrusion bioprinted shell-core hydrogel microfibers mimic glioma microenvironment and enhance the drug resistance of cancer cells. Colloids Surf B: Biointerfaces. 2018;171:291–9.

    Article  CAS  PubMed  Google Scholar 

  144. Pati F, Jang J, Ha D-H, Won Kim S, Rhie J-W, Shim J-H, Kim D-H, Cho D-W. Printing three-dimensional tissue analogues with decellularized extracellular matrix bioink. J Nature Commun. 2014;5(1):3935.

    Article  ADS  CAS  Google Scholar 

  145. Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nature Biotechnol. 2014;32(8):773–85.

    Article  CAS  Google Scholar 

  146. Datta P, Dey M, Ataie Z, Unutmaz D, Ozbolat IT. 3D bioprinting for reconstituting the cancer microenvironment. NPJ Precis Oncol. 2020;4(1):1–13.

    Google Scholar 

  147. Pandya HJ, Dhingra K, Prabhakar D, Chandrasekar V, Natarajan SK, Vasan AS, Kulkarni A, Shafiee H. A microfluidic platform for drug screening in a 3D cancer microenvironment. Biosens Bioelectr. 2017;94:632–42.

    Article  CAS  Google Scholar 

  148. Xu Z, Li E, Guo Z, Yu R, Hao H, Xu Y, Sun Z, Li X, Lyu J, Wang Q. Design and construction of a multi-organ microfluidic Chip mimicking the in vivo microenvironment of lung cancer metastasis. ACS Appl Mater Interfaces. 2016;8(39):25840–7.

    Article  CAS  PubMed  Google Scholar 

  149. Zhang YS, Zhang Y-N, Zhang W. Cancer-on-a-chip systems at the frontier of nanomedicine. Drug Discovery Today. 2017;22(9):1392–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Mi S, Du Z, Xu Y, Wu Z, Qian X, Zhang M, Sun W. Microfluidic co-culture system for cancer migratory analysis and anti-metastatic drugs screening. Sci Rep. 2016;6(1):35544.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  151. Kang L, Chung BG, Langer R, Khademhosseini A. Microfluidics for drug discovery and development: from target selection to product lifecycle management. Drug Discovery Today. 2008;13(1):1–13.

    Article  CAS  PubMed  Google Scholar 

  152. Papapetrou EP. Patient-derived induced pluripotent stem cells in cancer research and precision oncology. Nat Med. 2016;22(12):1392–401.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Menon NV, Lim SB, Lim CT. Microfluidics for personalized drug screening of cancer. Current opinion in pharmacology. 2019;48:155–61.

    Article  Google Scholar 

  154. Whitesides GM. The origins and the future of microfluidics. Nature. 2006;442(7101):368–73.

    Article  ADS  CAS  PubMed  Google Scholar 

  155. Albanese A, Lam AK, Sykes EA, Rocheleau JV, Chan WCW. Tumour-on-a-chip provides an optical window into nanoparticle tissue transport. Nat Commun. 2013;4(1):2718.

    Article  ADS  PubMed  Google Scholar 

  156. Sackmann EK, Fulton AL, Beebe DJ. The present and future role of microfluidics in biomedical research. Nature. 2014;507(7491):181–9.

    Article  ADS  CAS  PubMed  Google Scholar 

  157. Chen Y, Gao D, Liu H, Lin S, Jiang Y. Drug cytotoxicity and signaling pathway analysis with three-dimensional tumor spheroids in a microwell-based microfluidic chip for drug screening. Analytica Chimica Acta. 2015;898:85–92.

    Article  CAS  PubMed  Google Scholar 

  158. Shirure VS, Lezia A, Tao A, Alonzo LF, George SC. Low levels of physiological interstitial flow eliminate morphogen gradients and guide angiogenesis. Angiogenesis. 2017;20(4):493–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Fan Y, Nguyen DT, Akay Y, Xu F, Akay M. Engineering a brain cancer chip for high-throughput drug screening. Sci Rep. 2016;6:25062.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  160. Song JW, Cavnar SP, Walker AC, Luker KE, Gupta M, Tung Y-C, Luker GD, Takayama S. Microfluidic endothelium for studying the intravascular adhesion of metastatic breast cancer cells. PloS one. 2009;4(6): e5756.

    Article  ADS  PubMed  PubMed Central  Google Scholar 

  161. Sui W, Shi Z, Xue W, Ou M, Zhu Y, Chen J, Lin H, Liu F, Dai Y. Circular RNA and gene expression profiles in gastric cancer based on microarray chip technology. Oncol Rep. 2017;37(3):1804–14.

    Article  CAS  PubMed  Google Scholar 

  162. Gray JW, Mills GB. Large-scale drug screens support precision medicine. Cancer Discov. 2015;5(11):1130–2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  163. Shen W, Pei P, Zhang C, Li J, Han X, Liu T, Shi X, Su Z, Han G, Hu L, Yang K. A polymeric hydrogel to eliminate programmed death-ligand 1 for enhanced tumor radio-immunotherapy. ACS Nano. 2023;17(23):23998–4011.

    Article  CAS  PubMed  Google Scholar 

  164. Chen Y, Gao D, Wang Y, Lin S, Jiang Y. A novel 3D breast-cancer-on-chip platform for therapeutic evaluation of drug delivery systems. Analytica Chimica Actarnal. 2018;1036:97–106.

    Article  CAS  Google Scholar 

  165. Khazali AS, Clark AM, Wells A. A pathway to personalizing therapy for metastases using liver-on-a-Chip platforms. Stem Cell Rev Rep. 2017;13(3):364–80.

    Article  CAS  PubMed  Google Scholar 

  166. Hachey SJ, Hughes CC. Applications of tumor chip technology. Lab Chip. 2018;18(19):2893–912.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Hao S, Ha L, Cheng G, Wan Y, Xia Y, Sosnoski DM, Mastro AM, Zheng SY. A spontaneous 3D bone-on-a-chip for bone metastasis study of breast cancer cells. Small. 2018;14(12):1702787.

    Article  Google Scholar 

  168. Choi Y, Hyun E, Seo J, Blundell C, Kim HC, Lee E, Lee SH, Moon A, Moon WK, Huh D. A microengineered pathophysiological model of early-stage breast cancer. Lab Chip. 2015;15(16):3350–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346–54.

    Article  ADS  CAS  PubMed  Google Scholar 

  170. Fetah KL, DiPardo BJ, Kongadzem EM, Tomlinson JS, Elzagheid A, Elmusrati M, Khademhosseini A, Ashammakhi N. Cancer modeling-on-a-Chip with future artificial intelligence integration. Small. 2019;15(50):1901985.

    Article  CAS  Google Scholar 

  171. Bogorad MI, DeStefano J, Karlsson J, Wong AD, Gerecht S, Searson PC. Review: in vitro microvessel models. Lab on a Chip. 2015;15(22):4242–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Dereli-Korkut Z, Akaydin HD, Ahmed AR, Jiang X, Wang S. Three dimensional microfluidic cell arrays for ex vivo drug screening with mimicked vascular flow. Analyt Chem. 2014;86(6):2997–3004.

    Article  CAS  Google Scholar 

  173. Wang X-Y, Pei Y, Xie M, Jin Z-H, Xiao Y-S, Wang Y, Zhang L-N, Li Y, Huang W-H. An artificial blood vessel implanted three-dimensional microsystem for modeling transvascular migration of tumor cells. Lab Chip. 2015;15(4):1178–87.

    Article  CAS  PubMed  Google Scholar 

  174. Del Amo C, Borau C, Gutiérrez R, Asín J, García-Aznar JM. Quantification of angiogenic sprouting under different growth factors in a microfluidic platform. J Biomech. 2016;49(8):1340–6.

    Article  PubMed  Google Scholar 

  175. Nashimoto Y, Okada R, Hanada S, Arima Y, Nishiyama K, Miura T, Yokokawa R. Vascularized cancer on a chip: the effect of perfusion on growth and drug delivery of tumor spheroid. Biomaterials. 2020;229:119547.

    Article  CAS  PubMed  Google Scholar 

  176. Trietsch SJ, Israëls GD, Joore J, Hankemeier T, Vulto P. Microfluidic titer plate for stratified 3D cell culture. Lab Chip. 2013;13(18):3548–54.

    Article  CAS  PubMed  Google Scholar 

  177. Liu PF, Cao YW, Zhang SD, Zhao Y, Liu XG, Shi HQ, Hu KY, Zhu GQ, Ma B, Niu HT. A bladder cancer microenvironment simulation system based on a microfluidic co-culture model. Oncotarget. 2015;6(35):37695–705.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Yu T, Guo Z, Fan H, Song J, Liu Y, Gao Z, Wang Q. Cancer-associated fibroblasts promote non-small cell lung cancer cell invasion by upregulation of glucose-regulated protein 78 (GRP78) expression in an integrated bionic microfluidic device. Oncotarget. 2016;7(18):25593.

    Article  PubMed  PubMed Central  Google Scholar 

  179. Murrell M, Oakes PW, Lenz M, Gardel ML. Forcing cells into shape: the mechanics of actomyosin contractility. Nat Rev Mol Cell Biol. 2015;16(8):486–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Chin L, Xia Y, Discher DE, Janmey PA. Mechanotransduction in cancer. Curr Opin Chem Eng. 2016;11:77–84.

    Article  PubMed  PubMed Central  Google Scholar 

  181. Mitchell MJ, King MR. Computational and experimental models of cancer cell response to fluid shear stress. Front Oncol. 2013;3:44.

    Article  PubMed  PubMed Central  Google Scholar 

  182. Swartz MA, Lund AW. Lymphatic and interstitial flow in the tumour microenvironment: linking mechanobiology with immunity. Nat Rev Cancer. 2012;12(3):210–9.

    Article  CAS  PubMed  Google Scholar 

  183. Ip CK, Li S-S, Tang MY, Sy SK, Ren Y, Shum HC, Wong AS. Stemness and chemoresistance in epithelial ovarian carcinoma cells under shear stress. Sci Rep. 2016;6:26788.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  184. Muz B, de la Puente P, Azab F, Azab AK. The role of hypoxia in cancer progression, angiogenesis, metastasis, and resistance to therapy. Hypoxia (Auckl). 2015;3:83–92.

    Article  PubMed  Google Scholar 

  185. Wilson WR, Hay MP. Targeting hypoxia in cancer therapy. Nature Reviews Cancer. 2011;11(6):393–410.

    Article  CAS  PubMed  Google Scholar 

  186. Ramesan S, Rezk AR, Cheng KW, Chan PP, Yeo LY. Acoustically-driven thread-based tuneable gradient generators. Lab Chip. 2016;16(15):2820–8.

    Article  CAS  PubMed  Google Scholar 

  187. Wang H, Chen C-H, Xiang Z, Wang M, Lee C. A convection-driven long-range linear gradient generator with dynamic control. Lab Chip. 2015;15(6):1445–50.

    Article  PubMed  Google Scholar 

  188. Kamei K-I, Mashimo Y, Koyama Y, Fockenberg C, Nakashima M, Nakajima M, Li J, Chen Y. 3D printing of soft lithography mold for rapid production of polydimethylsiloxane-based microfluidic devices for cell stimulation with concentration gradients. Biomed Microdevices. 2015;17(2):36.

    Article  PubMed  Google Scholar 

  189. Zou H, Yue W, Yu W-K, Liu D, Fong C-C, Zhao J, Yang M. Microfluidic platform for studying chemotaxis of adhesive cells revealed a gradient-dependent migration and acceleration of cancer stem cells. Analyt Chem. 2015;87(14):7098–108.

    Article  CAS  Google Scholar 

  190. Ehsan SM, George SC. Vessel network formation in response to intermittent hypoxia is frequency dependent. J Biosci Bioeng. 2015;120(3):347–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  191. Acosta MA, Jiang X, Huang P-K, Cutler KB, Grant CS, Walker GM, Gamcsik MP. A microfluidic device to study cancer metastasis under chronic and intermittent hypoxia. J Biomicrofluidics. 2014;8(5):054117.

    Article  Google Scholar 

  192. Aung A, Theprungsirikul J, Lim HL, Varghese S. Chemotaxis-driven assembly of endothelial barrier in a tumor-on-a-chip platform. Lab Chip. 2016;16(10):1886–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Tahara RK, Brewer TM, Theriault RL, Ueno NT. Bone metastasis of breast cancer. Breast Cancer Metastasis Drug Resist: Challenges Progress 201;1152:105–29. https://doi.org/10.1007/978-3-030-20301-6_7.

  194. Zhu W, Wang M, Fu Y, Castro NJ, Fu SW, Zhang LG. Engineering a biomimetic three-dimensional nanostructured bone model for breast cancer bone metastasis study. Acta biomaterialia. 2015;14:164–74.

    Article  ADS  CAS  PubMed  Google Scholar 

  195. Zhang Y, Ma B, Fan Q. Mechanisms of breast cancer bone metastasis. Cancer letters. 2010;292(1):1–7.

    Article  ADS  CAS  PubMed  Google Scholar 

  196. Wright LE, Ottewell PD, Rucci N, Peyruchaud O, Pagnotti GM, Chiechi A, Buijs JT, Sterling JA. Murine models of breast cancer bone metastasis. Bone Key Rep. 2016;5:804. https://doi.org/10.1038/bonekey.2016.31.

  197. Simmons J, Hildreth B III, Supsavhad W, Elshafae S, Hassan B, Dirksen W, Toribio RE, Rosol TJ. Animal models of bone metastasis. Vet Pathol. 2015;52(5):827–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  198. Lefley D, Howard F, Arshad F, Bradbury S, Brown H, Tulotta C, Eyre R, Alférez D, Wilkinson JM, Holen I. Development of clinically relevant in vivo metastasis models using human bone discs and breast cancer patient-derived xenografts. Breast Cancer Res. 2019;21(1):1–21.

    Article  Google Scholar 

  199. Ireson CR, Alavijeh MS, Palmer AM, Fowler ER, Jones HJ. The role of mouse tumour models in the discovery and development of anticancer drugs. Brit J Cancer. 2019;121(2):101–8.

    Article  PubMed  PubMed Central  Google Scholar 

  200. Manning HC, Buck JR, Cook RS. Mouse models of breast cancer: platforms for discovering precision imaging diagnostics and future cancer medicine. J Nuclear Med. 2016;57(Supplement 1):60S-68S.

    Article  Google Scholar 

  201. Chen S, Zeng J, Huang L, Peng Y, Yan Z, Zhang A, Zhao X, Li J, Zhou Z, Wang S, Jing S, Hu M, Li Y, Wang D, Wang W, Yu H, Miao J, Li J, Deng Y, Li Y, Liu T, Xu D. RNA adenosine modifications related to prognosis and immune infiltration in osteosarcoma. J Transl Med. 2022;20(1):228.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Olson B, Li Y, Lin Y, Liu ET, Patnaik A. Mouse models for cancer immunotherapy ResearchCoclinical mouse models for cancer immunotherapy. Cancer Discov. 2018;8(11):1358–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  203. Park MK, Lee CH, Lee H. Mouse models of breast cancer in preclinical research. Labor Anim Res. 2018;34:160–5.

    Article  Google Scholar 

  204. Nakayama J, Han Y, Kuroiwa Y, Azuma K, Yamamoto Y, Semba K. The in vivo selection method in breast cancer metastasis. Int J Mol Sci. 2021;22(4):1886.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  205. Holen I, Speirs V, Morrissey B, Blyth K. In vivo models in breast cancer research: progress, challenges and future directions. Dis Models Mechan. 2017;10(4):359–71.

    Article  CAS  Google Scholar 

  206. Lelekakis M, Moseley JM, Martin TJ, Hards D, Williams E, Ho P, Lowen D, Javni J, Miller FR, Slavin J. A novel orthotopic model of breast cancer metastasis to bone. Clin Exper Metastasis. 1999;17:163–70.

    Article  CAS  Google Scholar 

  207. Lee J-H, Kim B, Jin WJ, Kim J-W, Kim H-H, Ha H, Lee ZH. Trolox inhibits osteolytic bone metastasis of breast cancer through both PGE2-dependent and independent mechanisms. Biochem Pharmacol. 2014;91(1):51–60.

    Article  ADS  CAS  PubMed  Google Scholar 

  208. Farhoodi HP, Segaliny AI, Wagoner ZW, Cheng JL, Liu L, Zhao W. Optimization of a syngeneic murine model of bone metastasis. J Bone Oncol. 2020;23:100298.

    Article  PubMed  PubMed Central  Google Scholar 

  209. Han Y, Nakayama J, Hayashi Y, Jeong S, Futakuchi M, Ito E, Watanabe S, Semba K. Establishment and characterization of highly osteolytic luminal breast cancer cell lines by intracaudal arterial injection. Genes Cells. 2020;25(2):111–23.

    Article  CAS  PubMed  Google Scholar 

  210. Gómez-Cuadrado L, Tracey N, Ma R, Qian B, Brunton VG. Mouse models of metastasis: progress and prospects. Dis Models Mechanisms. 2017;10(9):1061–74.

    Article  Google Scholar 

  211. Yoneda T, Sasaki A, Dunstan C, Williams PJ, Bauss F, De Clerck YA, Mundy GR. Inhibition of osteolytic bone metastasis of breast cancer by combined treatment with the bisphosphonate ibandronate and tissue inhibitor of the matrix metalloproteinase-2. J Clin Invest. 1997;99(10):2509–17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Yi B, Williams PJ, Niewolna M, Wang Y, Yoneda T. Tumor-derived platelet-derived growth factor-BB plays a critical role in osteosclerotic bone metastasis in an animal model of human breast cancer. Cancer Res. 2002;62(3):917–23.

    CAS  PubMed  Google Scholar 

  213. Ottewell PD, Wang N, Brown HK, Reeves KJ, Fowles CA, Croucher PI, Eaton CL, Holen I. Zoledronic acid has differential antitumor activity in the pre-and postmenopausal bone microenvironment in vivo. Clin Cancer Res. 2014;20(11):2922–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  214. Suva LJ, Washam C, Nicholas RW, Griffin RJ. Bone metastasis: mechanisms and therapeutic opportunities. Nat Rev Endocrinol. 2011;7(4):208–18.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  215. Wetterwald A, van der Pluijm G, Que I, Sijmons B, Buijs J, Karperien M, Löwik CW, Gautschi E, Thalmann GN, Cecchini MG. Optical imaging of cancer metastasis to bone marrow: a mouse model of minimal residual disease. Am J Pathol. 2002;160(3):1143–53.

    Article  PubMed  PubMed Central  Google Scholar 

  216. Holen I, Nutter F, Wilkinson J, Evans C, Avgoustou P, Ottewell PD. Human breast cancer bone metastasis in vitro and in vivo: a novel 3D model system for studies of tumour cell-bone cell interactions. Clin Exper Metastasis. 2015;32:689–702.

    Article  CAS  Google Scholar 

  217. Kuperwasser C, Dessain S, Bierbaum BE, Garnet D, Sperandio K, Gauvin GP, Naber SP, Weinberg RA, Rosenblatt M. A mouse model of human breast cancer metastasis to human bone. Cancer Res. 2005;65(14):6130–8.

    Article  CAS  PubMed  Google Scholar 

  218. Werbeck J, Thudi N, Martin C, Premanandan C, Yu L, Ostrowksi M, Rosol T. Tumor microenvironment regulates metastasis and metastasis genes of mouse MMTV-PymT mammary cancer cells in vivo. Veterinary pathology. 2014;51(4):868–81.

    Article  CAS  PubMed  Google Scholar 

  219. Ottewell P, Coleman R, Holen I. From genetic abnormality to metastases: murine models of breast cancer and their use in the development of anticancer therapies. Breast Cancer Res Treatment. 2006;96:101–13.

    Article  CAS  Google Scholar 

  220. Chulpanova DS, Kitaeva KV, Rutland CS, Rizvanov AA, Solovyeva VV. Mouse tumor models for advanced cancer immunotherapy. Int J Mol Sci. 2020;21(11):4118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  221. Brekke TD, Steele KA, Mulley JF. Inbred or outbred? Genetic diversity in laboratory rodent colonies. G3 Genes Genomes Genet. 2018;8(2):679–86.

    Article  CAS  Google Scholar 

  222. Cespedes MV, Casanova I, Parreño M, Mangues R. Mouse models in oncogenesis and cancer therapy. Clin Transl Oncol. 2006;8:318–29.

    Article  CAS  PubMed  Google Scholar 

  223. Nandi S, Guzman RC, Yang J. Hormones and mammary carcinogenesis in mice, rats, and humans: a unifying hypothesis. Proc Natl Acad Sci. 1995;92(9):3650–7.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  224. Buijs JT, Matula KM, Cheung H, Kruithof-de Julio M, Van Der Mark MH, Snoeks TJ, Cohen R, Corver WE, Mohammad KS, Jonkers J. Spontaneous bone metastases in a preclinical orthotopic model of invasive lobular carcinoma; the effect of pharmacological targeting TGFβ receptor I kinase. J Pathol. 2015;235(5):745–59.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  225. Pulaski BA, Ostrand-Rosenberg S. Mouse 4T1 breast tumor model. Curr Prot Immun. 2000;39(1):20.2.1-20.2.16.

    Google Scholar 

  226. Tulotta C, Groenewoud A, Snaar-Jagalska BE, Ottewell P. Animal models of breast cancer bone metastasis. Bone Res Protoc. Methods Mol Biol. 2019:1914:309–30. https://doi.org/10.1007/978-1-4939-8997-3_17.

  227. Reed ND, Manning DD. Long-term maintenance of normal human skin on congenitally athymic (nude) mice. Proc Soc Exper Biol Med. 1973;143(2):350–3.

    Article  CAS  Google Scholar 

  228. Kang Y, Siegel PM, Shu W, Drobnjak M, Kakonen SM, Cordón-Cardo C, Guise TA, Massagué J. A multigenic program mediating breast cancer metastasis to bone. Cancer Cell. 2003;3(6):537–49.

    Article  CAS  PubMed  Google Scholar 

  229. Holen I, Walker M, Nutter F, Fowles A, Evans C, Eaton C, Ottewell P. Oestrogen receptor positive breast cancer metastasis to bone: inhibition by targeting the bone microenvironment in vivo. Clin Exper Metastasis. 2016;33:211–24.

    Article  CAS  Google Scholar 

  230. Zhang X, Lewis MT. Establishment of patient-derived xenograft (PDX) models of human breast cancer. Curr Protoc Mouse Biol. 2013;3(1):21–9.

    Article  PubMed  Google Scholar 

  231. Wei S, Sun T, Du J, Zhang B, Xiang D, Li W. Xanthohumol, a prenylated flavonoid from hops, exerts anticancer effects against gastric cancer in vitro. Oncol Rep. 2018;40(6):3213–22.

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Aliabadi A, Hasanvand Z, Kiani A, Mirabdali SS. Synthesis and in-vitro cytotoxicity assessment of N-(5-(benzylthio)-1,3,4- thiadiazol-2-yl)-2-(4-(trifluoromethyl)phenyl)acetamide with potential anticancer activity. Iran J Pharm Res. 2013;12(4):687–93.

    CAS  PubMed  PubMed Central  Google Scholar 

  233. Tulotta C, He S, Van Der Ent W, Chen L, Groenewoud A, Spaink H, Snaar-Jagalska B. Imaging cancer angiogenesis and metastasis in a zebrafish embryo model. Cancer and Zebrafish. Advances in Experimental Medicine and Biology 2016;916:239–63. https://doi.org/10.1007/978-3-319-30654-4_11.

  234. Mercatali L, La Manna F, Groenewoud A, Casadei R, Recine F, Miserocchi G, Pieri F, Liverani C, Bongiovanni A, Spadazzi C. Development of a patient-derived xenograft (PDX) of breast cancer bone metastasis in a zebrafish model. Int J Mol Sci. 2016;17(8):1375.

    Article  PubMed  PubMed Central  Google Scholar 

  235. Chen X, Li Y, Yao T, Jia R. Benefits of zebrafish xenograft models in cancer research. Front Cell Dev Biol. 2021;9:616551.

    Article  PubMed  PubMed Central  Google Scholar 

  236. Chernet DY, Klassen L, Goertzen S, Malagon JN. Live imaging and quantification of circulating potentially metastatic tumor cells in early pupal stage of Drosophila melanogaster. Micropubl Biol. 2022; https://doi.org/10.17912/micropub.biology.000588.

  237. Kirschbaum A, Geisse NC, Sister TJ, Meyer LM. Effect of certain folic acid antagonists on transplanted myeloid and lymphoid leukemias of the F strain of mice. Cancer Res. 1950;10(12):762–8.

    CAS  PubMed  Google Scholar 

  238. Gazdar AF, Hirsch FR, Minna JD. From mice to men and back: an assessment of preclinical model systems for the study of lung cancers. J Thor Oncol. 2016;11(3):287–99.

    Article  Google Scholar 

  239. Hutchinson L, Kirk R. High drug attrition rates—where are we going wrong? Nat Rev Clin Oncol. 2011;8(4):189–90.

    Article  PubMed  Google Scholar 

  240. Wang Y, Cui J, Wang L. Patient-derived xenografts: a valuable platform for clinical and preclinical research in pancreatic cancer. Chin Clin Oncol. 2019;8:2 Chinese Clinical Oncology (Genetic Features of Pancreatic Cancer).

    Article  Google Scholar 

  241. Koga Y, Ochiai A. Systematic review of patient-derived xenograft models for preclinical studies of anti-cancer drugs in solid tumors. Cells. 2019;8(5):418.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  242. Hidalgo M, Amant F, Biankin AV, Budinska E, Byrne AT, Caldas C, Clarke RB, de Jong S, Jonkers J, Maelandsmo GM, Roman-Roman S, Seoane J, Trusolino L, Villanueva A. Patient-derived xenograft models: an emerging platform for translational cancer research. J Cancer Discov. 2014;4(9):998–1013.

    Article  CAS  Google Scholar 

  243. Zhao X, Liu Z, Yu L, Zhang Y, Baxter P, Voicu H, Gurusiddappa S, Luan J, Su JM, Leung HC, Li XN. Global gene expression profiling confirms the molecular fidelity of primary tumor-based orthotopic xenograft mouse models of medulloblastoma. Neuro Oncol. 2012;14(5):574–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  244. Lawson DA, Bhakta NR, Kessenbrock K, Prummel KD, Yu Y, Takai K, Zhou A, Eyob H, Balakrishnan S, Wang CY, Yaswen P, Goga A, Werb Z. Single-cell analysis reveals a stem-cell program in human metastatic breast cancer cells. Nature. 2015;526(7571):131–5.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  245. Evans KW, Yuca E, Akcakanat A, Scott SM, Arango NP, Zheng X, Chen K, Tapia C, Tarco E, Eterovic AK, Black DM, Litton JK, Yap TA, Tripathy D, Mills GB, Meric-Bernstam F. A population of heterogeneous breast cancer patient-derived xenografts demonstrate broad activity of PARP inhibitor in BRCA1/2 wild-type tumors. Clin Cancer Res. 2017;23(21):6468–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  246. Yu J, Qin B, Moyer AM, Nowsheen S, Liu T, Qin S, Zhuang Y, Liu D, Lu SW, Kalari KR, Visscher DW, Copland JA, McLaughlin SA, Moreno-Aspitia A, Northfelt DW, Gray RJ, Lou Z, Suman VJ, Weinshilboum R, Boughey JC, Goetz MP, Wang L. DNA methyltransferase expression in triple-negative breast cancer predicts sensitivity to decitabine. J Clin Invest. 2018;128(6):2376–88.

    Article  PubMed  PubMed Central  Google Scholar 

  247. Xiao T, Li W, Wang X, Xu H, Yang J, Wu Q, Huang Y, Geradts J, Jiang P, Fei T, Chi D, Zang C, Liao Q, Rennhack J, Andrechek E, Li N, Detre S, Dowsett M, Jeselsohn RM, Liu XS, Brown M. Estrogen-regulated feedback loop limits the efficacy of estrogen receptor-targeted breast cancer therapy. Proc Natl Acad Sci USA. 2018;115(31):7869–78.

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  248. El Ayachi I, Fatima I, Wend P, Alva-Ornelas JA, Runke S, Kuenzinger WL, Silva J, Silva W, Gray JK, Lehr S, Barch HC, Krutilina RI, White AC, Cardiff R, Yee LD, Yang L, O’Regan RM, Lowry WE, Seagroves TN, Seewaldt V, Krum SA, Miranda-Carboni GA. The WNT10B network is associated with survival and metastases in chemoresistant triple-negative breast cancer. Cancer Res. 2019;79(5):982–93.

    Article  PubMed  Google Scholar 

  249. Reyal F, Guyader C, Decraene C, Lucchesi C, Auger N, Assayag F, De Plater L, Gentien D, Poupon MF, Cottu P, De Cremoux P, Gestraud P, Vincent-Salomon A, Fontaine JJ, Roman-Roman S, Delattre O, Decaudin D, Marangoni E. Molecular profiling of patient-derived breast cancer xenografts. Breast Cancer Res. 2012;14(1):R11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  250. George E, Kim H, Krepler C, Wenz B, Makvandi M, Tanyi JL, Brown E, Zhang R, Brafford P, Jean S, Mach RH, Lu Y, Mills GB, Herlyn M, Morgan M, Zhang X, Soslow R, Drapkin R, Johnson N, Zheng Y, Cotsarelis G, Nathanson KL, Simpkins F. A patient-derived-xenograft platform to study BRCA-deficient ovarian cancers. JCI Insight. 2017;2(1): e89760.

    Article  PubMed  PubMed Central  Google Scholar 

  251. Nunes M, Vrignaud P, Vacher S, Richon S, Lievre A, Cacheux W, Weiswald L-B, Massonnet G, Chateau-Joubert S, Nicolas A. Evaluating patient-derived colorectal cancer xenografts as preclinical models by comparison with patient clinical data. Cancer research. 2015;75(8):1560–6.

    Article  CAS  PubMed  Google Scholar 

  252. Clevers H. Modeling development and disease with organoids. Cell. 2016;165(7):1586–97.

    Article  CAS  PubMed  Google Scholar 

  253. Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, Sailer V, Augello M, Puca L, Rosati R, McNary TJ, Churakova Y, Cheung C, Triscott J, Pisapia D, Rao R, Mosquera JM, Robinson B, Faltas BM, Emerling BE, Gadi VK, Bernard B, Elemento O, Beltran H, Demichelis F, Kemp CJ, Grandori C, Cantley LC, Rubin MA. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 2017;7(5):462–77.

    Article  PubMed  PubMed Central  Google Scholar 

  254. Lu P, Takai K, Weaver VM, Werb Z. Extracellular matrix degradation and remodeling in development and disease. Cold Spring Harb Perspect Biol. 2011;3(12):a005058.

    Article  PubMed  PubMed Central  Google Scholar 

  255. DeRose YS, Wang G, Lin YC, Bernard PS, Buys SS, Ebbert MT, Factor R, Matsen C, Milash BA, Nelson E, Neumayer L, Randall RL, Stijleman IJ, Welm BE, Welm AL. Tumor grafts derived from women with breast cancer authentically reflect tumor pathology, growth, metastasis and disease outcomes. Nat Med. 2011;17(11):1514–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  256. Clohessy JG, Pandolfi PP. Mouse hospital and co-clinical trial project—from bench to bedside. Nat Rev Clin Oncol. 2015;12(8):491.

    Article  PubMed  Google Scholar 

  257. Williams JA. Using PDX for preclinical cancer drug discovery: the evolving field. J Clin Med. 2018;7(3):41.

    Article  PubMed  PubMed Central  Google Scholar 

  258. Inoue A, Deem AK, Kopetz S, Heffernan TP, Draetta GF, Carugo A. Current and future horizons of patient-derived xenograft models in colorectal cancer translational research. Cancers (Basel). 2019;11(9):1321.

    Article  CAS  PubMed  Google Scholar 

  259. Pearson AT, Finkel KA, Warner KA, Nör F, Tice D, Martins MD, Jackson TL, Nör JE. Patient-derived xenograft (PDX) tumors increase growth rate with time. Oncotarget 2016;7(7):7993–8005; https://doi.org/10.18632/oncotarget.6919.

  260. Wang Y, Cui J, Wang L. Patient-derived xenografts: a valuable platform for clinical and preclinical research in pancreatic cancer. Chin Clin Oncol. 2019;8(2):17.

    Article  PubMed  Google Scholar 

  261. Wang X, Chen G, Zhang Y, Ghareeb WM, Yu Q, Zhu H, Lu X, Huang Y, Huang S, Hou D, Chi P. The impact of circumferential tumour location on the clinical outcome of rectal cancer patients managed with neoadjuvant chemoradiotherapy followed by total mesorectal excision. Eur J Surg Oncol. 2020;46(6):1118–23.

    Article  PubMed  Google Scholar 

  262. Hoffman RM. Patient-derived orthotopic xenografts: better mimic of metastasis than subcutaneous xenografts. Nat Rev Cancer. 2015;15(8):451–2.

    Article  CAS  PubMed  Google Scholar 

  263. Liang M, Zhang P, Fu J. Up-regulation of LOX-1 expression by TNF-α promotes trans-endothelial migration of MDA-MB-231 breast cancer cells. Cancer letters. 2007;258(1):31–7.

    Article  CAS  PubMed  Google Scholar 

  264. Pignatelli J, Goswami S, Jones JG, Rohan TE, Pieri E, Chen X, Adler E, Cox D, Maleki S, Bresnick A. Invasive breast carcinoma cells from patients exhibit MenaINV-and macrophage-dependent transendothelial migration. Sci Signaling. 2014;7(353):ra112.

    Article  Google Scholar 

  265. Group EBCTC. Adjuvant bisphosphonate treatment in early breast cancer: meta-analyses of individual patient data from randomised trials. Lancet. 2015;386(10001):1353–61.

    Article  Google Scholar 

  266. Smalley KS, Lioni M, Noma K, Haass NK, Herlyn M. In vitro three-dimensional tumor microenvironment models for anticancer drug discovery. Expert Opin Drug Discov. 2008;3(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  267. Nugraha B, Hong X, Mo X, Tan L, Zhang W, Chan P-M, Kang CH, Wang Y, Beng LT, Sun W. Galactosylated cellulosic sponge for multi-well drug safety testing. Biomaterials. 2011;32(29):6982–94.

    Article  CAS  PubMed  Google Scholar 

  268. Bahcecioglu G, Basara G, Ellis BW, Ren X, Zorlutuna P. Breast cancer models: engineering the tumor microenvironment. Acta biomaterialia. 2020;106:1–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  269. Rejniak KA. Investigating dynamical deformations of tumor cells in circulation: predictions from a theoretical model. Front Oncol. 2012;2:111.

    Article  PubMed  PubMed Central  Google Scholar 

  270. Baskaran JP, Weldy A, Guarin J, Munoz G, Shpilker PH, Kotlik M, Subbiah N, Wishart A, Peng Y, Miller MA. Cell shape, and not 2D migration, predicts extracellular matrix-driven 3D cell invasion in breast cancer. APL Bioeng. 2020;4(2):026105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  271. Faiella E, Santucci D, Calabrese A, Russo F, Vadalà G, Zobel BB, Soda P, Iannello G, de Felice C, Denaro V. Artificial intelligence in bone metastases: an MRI and CT imaging review. Int J Environ Res Publ Health. 2022;19(3):1880.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge the Deputy for Research and Technology, Tabriz University of Medical Sciences, for grant support (73668).

Funding

This work was funded by Deputy for Research and Technology, Tabriz University of Medical Sciences [grant number: 73668].

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the investigation, conceptualization, and analysis of the information in this manuscript, and were involved in the writing process.

Corresponding authors

Correspondence to Nima Beheshtizadeh or Nima Rezaei.

Ethics declarations

Ethics approval and consent to participate

This article does not contain any studies with human participants or animals performed by any of the authors.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kolahi Azar, H., Gharibshahian, M., Rostami, M. et al. The progressive trend of modeling and drug screening systems of breast cancer bone metastasis. J Biol Eng 18, 14 (2024). https://doi.org/10.1186/s13036-024-00408-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13036-024-00408-5

Keywords