Next Article in Journal
Factor XII—A New Therapeutic Target? A Systematic Review
Previous Article in Journal
Possible Impact of Lymphatic Drainage on Brain Injury After Aneurysmal Subarachnoid Hemorrhage
Previous Article in Special Issue
Organ-on-a-Chip: A Roadmap for Translational Research in Human and Veterinary Medicine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Mechano-Organ-on-Chip for Cancer Research

1
School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
2
Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
3
Department of Biomedical Engineering, Faculty of Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China
4
Department of Biomedical Engineering, College of Biomedicine, City University of Hong Kong, Kowloon, Hong Kong SAR, China
5
Hong Kong Centre of Cerebro-Cardiovascular Health Engineering (COCHE), Shatin, Hong Kong SAR, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2026, 27(3), 1330; https://doi.org/10.3390/ijms27031330 (registering DOI)
Submission received: 20 December 2025 / Revised: 20 January 2026 / Accepted: 24 January 2026 / Published: 29 January 2026
(This article belongs to the Special Issue Organoids and Organs-on-Chip for Medical Research)

Abstract

Mechano-Organ-on-Chip (Mechano-OoC) platforms are emerging as powerful microphysiological systems that place mechanical cues at the center of tumor modeling, providing a scalable and human-relevant approach to recapitulate the biophysical complexity of the tumor microenvironment. Mechanical factors such as matrix stiffness, viscoelasticity, solid stress, interstitial flow, confinement, and shear critically regulate cancer progression, metastasis, immune interactions, and treatment response, yet remain poorly captured by conventional in vitro models and are often studied separately in tumor-on-chip and mechanobiology research. In this review, we summarize recent advances in mechano-OoC technologies for cancer research, highlighting strategies that integrate engineered mechanical cues with microfluidics, tunable extracellular matrices, vascular and stromal interfaces, and dynamic loading to model tumor invasion, vascular transport, immune trafficking, and drug delivery. We also discuss emerging approaches for real-time, multimodal readouts, including sensor-integrated platforms and artificial intelligence-assisted data analysis, and outline key challenges that limit translation, such as device complexity, limited throughput, insufficient standardization, and inadequate validation against in vivo and clinical data. By organizing progress across platform engineering, sensing and readout, data standardization, and AI-driven analytics, this review provides a unified framework for advancing mechanobiology-aware tumor models and guiding the development of predictive preclinical platforms for precision cancer therapy.

1. Introduction

It is increasingly recognized that the tumor microenvironment (TME) is governed by not only biochemical factors but also by physical and mechanical cues. Examples of such mechanical factors include extracellular matrix (ECM) stiffness, compressive solid stress, interstitial fluid pressure/flow, geometric confinement, and shear forces [1,2,3]. These forces influence key cancer phenotypes, including cell proliferation, survival, motility/invasion, epithelial–mesenchymal transition (EMT), stemness, and drug resistance [4]. Mechanotransduction, which is the process by which cancer cells sense these mechanical signals via the cytoskeleton, adhesion complexes, and nuclear deformation, can rewire gene expression and alter cellular behavior [5]. Moreover, non-malignant TME components and the ECM itself respond to such mechanical cues, affecting tumor progression, metastasis, immune interactions, and drug delivery [6,7]. Collectively, we emphasize that faithful tumor models must reproduce both the biochemical and mechanical dimensions of the TME.
Conventional preclinical models have major limitations in recapitulating tumor mechanics. Two-dimensional cell cultures entirely lack three-dimensional architecture, native ECM, fluid flow, and relevant mechanical forces [8]. Even advanced three-dimensional models, such as multicellular spheroids or organoids, which capture some cell–cell interactions, generally lack perfused vasculature, interstitial shear stress, stromal and immune components, and fail to reproduce the mechanical stresses experienced in vivo [9,10,11,12]. Animal models incorporate physiological complexity but introduce species-specific differences in ECM composition and immune context, incur high cost and ethical concerns, and limit high-throughput experimentation and real-time monitoring [13]. Consequently, there is a critical need for scalable, human-relevant in vitro platforms that integrate biochemical, cellular, and mechanical complexity [14,15,16]. We therefore propose that mechanobiology-aware Organ-on-Chip (Mechano-OoC) systems, which combine microfluidics, tissue engineering, and controlled mechanical cues, offer a promising solution to bridge this gap.
In this review, we highlight how OoC technology leverages microfabrication, microfluidics, and engineered biomaterials to create microphysiological systems that recapitulate key organ-level functions, incorporating controlled fluid flow, mechanical forces, multiple tissue interfaces, and three-dimensional architecture with human cells [17,18,19]. Recent studies emphasize that microfluidic OoC models can recreate tumor-like microenvironments while enabling precise tuning of biochemical and biophysical parameters. Given the critical role of mechanical forces in driving cancer invasion, metastasis, drug transport, and immune interactions, we focus on the growing need to integrate mechanobiology into OoC platforms; mechano-OoC systems, by incorporating engineered mechanical cues into tumor-on-chip models, offer a promising solution to achieve more realistic replication of these processes [20,21,22]. We summarize recent advances in engineering these platforms (including tunable ECM mechanics, dynamic loading, and multicellular interfaces), multimodal sensing and real-time readouts (with sensor integration), data standardization, and AI-driven image analysis and predictive modeling, while addressing challenges in scalability, reproducibility, and clinical validation to advance mechano-OoC as predictive tools for precision cancer therapy [23,24,25].

2. Engineering Mechano-OoC Platforms: Recapitulating the Mechanical Tumor Microenvironment

2.1. Key Mechanical Dimensions in the Tumor Microenvironment

Several key mechanical features characterize solid tumors. A central factor is the extracellular matrix: many tumors exhibit increased ECM cross-linking, fiber alignment, and density, yielding higher stiffness [26,27,28]. Changes in matrix viscoelasticity also modulate cell mechanosensing and behavior [29,30]. Another hallmark is growth-induced solid stress: as tumors expand and deposit ECM, compressive stresses build up that physically squeeze adjacent tissues and vessels, altering interstitial pressure and perfusion [31,32]. The geometry of the ECM, which includes features like pore size, fiber orientation, and physical confinement, further influences how cells migrate and how their nuclei deform under stress [33,34]. Importantly, the TME is heterogeneous and dynamically remodels over time: for example, matrix stiffness and architecture change through ECM deposition, degradation, and cell-driven remodeling [35,36,37]. These mechanical factors interact (e.g., ECM stiffness affects stress distribution, and fluid flow depends on matrix porosity), suggesting that mechano-OoC platforms should incorporate multiple cues simultaneously rather than focusing on a single parameter [38,39].
The competitive advantage of mechano-OoC platforms is not merely the ability to present multiple mechanical cues, but the capacity of microfluidic engineering to decouple those cues so they can be varied independently and quantitatively. Classic multi-layer designs use isolated vacuum or pneumatic drive channels that deform a thin membrane or hydrogel layer to apply cyclic stretch while keeping perfusion streams separate, enabling stretch amplitudes and frequencies to be changed without altering channel flow fields [40]. This architecture was pioneered in lung-on-a-chip devices and remains a canonical example of independent actuation.
Likewise, hydrogels or 3D matrices can be sandwiched or spatially confined in dedicated gel chambers that are fluidically isolated from adjacent perfusion channels. By imposing a controlled pressure difference across the gel or by perfusing parallel side channels, researchers can generate interstitial flow through the matrix while maintaining a low shear stress on cells at the matrix–channel interface [41]. Such geometries permit independent modulation of interstitial velocity and luminal shear stress. Beyond static geometry, tunable materials and active hydrogel strategies offer another route to decoupling: phototunable or chemically triggered hydrogels and actuatable hydrogel membranes allow in-platform changes in local matrix stiffness without changing external flow conditions, so stiffness-dependent mechanotransduction can be probed while holding convective transport constant [42].

2.2. Mechano-OoC Design Principles and Material Selection

General guidelines for Organ-on-Chip design have been articulated in recent research. To develop OoC systems, it is essential to start with a strong baseline that encompasses critical elements such as suitable materials, standardized cell seeding approaches, regulated microfluidic flow, and reliable readout strategies [43,44]. Extending these principles to cancer models involves adding modules for mechanical control. The same microfabrication and fluidic toolkits can be used, but the design must explicitly incorporate tunable ECM mechanics, on-chip force application, integrated sensors, and means for dynamic perturbation. Using collagen-gelatin composite hydrogels (with a tunable stiffness of 1–10 kPa) enables the accurate reproduction of the high-stiffness microenvironment of the pancreatic cancer stroma, thereby providing a controllable model for studying the matrix stiffness-induced EMT process [39,45].
A major technical limitation in tumor-on-chip platforms for drug screening lies in the non-specific adsorption and absorption of hydrophobic small-molecule drugs by polydimethylsiloxane (PDMS). Owing to its hydrophobic and porous polymer network, PDMS can significantly deplete the free concentration of many chemotherapeutic agents from the perfusion medium in a time- and compound-dependent manner [46]. This effect is particularly problematic in mechano-OoC systems, where long-term perfusion and cyclic mechanical stimulation can exacerbate drug partitioning into the PDMS bulk, thereby distorting the intended dose, exposure kinetics, and reproducibility of pharmacological readouts. Consequently, the apparent cellular response may reflect material–drug interactions rather than true mechano-pharmacological effects.
To mitigate these issues, surface coatings or PDMS pre-saturation strategies have been explored, yet these approaches often provide incomplete or unstable suppression of sorption and may compromise gas permeability or mechanical compliance under repeated deformation. As a result, alternative materials are increasingly adopted for quantitative drug screening applications. Thermoplastic polymers such as polystyrene (PS), poly methyl methacrylate (PMMA), and cyclic olefin copolymer/polymer (COC/COP) exhibit substantially reduced small-molecule sorption and improved chemical fidelity [47]. However, their limited elasticity necessitates hybrid or composite device designs, in which rigid thermoplastic microfluidic layers are combined with thin elastomeric or thermoplastic elastomer membranes to preserve mechanical actuation. Material selection thus represents a critical design parameter in mechano-OoC systems, particularly when accurate drug concentration control under dynamic mechanical loading is required for translational relevance.

2.3. Existing Mechano-OoC Platform Implementations and Applications

Several recent publications illustrate the promise of tumor-on-chip systems in cancer research. These current studies aim to establish a platform that can reproduce the significant characteristics of tumors, including three-dimensional structure, natural extracellular matrix, perfusion, heterogeneity, and even angiogenesis, for the purposes of cancer biology, metastasis, and drug testing studies (Figure 1A) [48,49]. More recent experiments have co-cultured tumor cells with stromal cells in microfluidic chips, modeling aspects of solid tumor growth under well-controlled microenvironmental conditions (Figure 1B) [50,51]. Similarly, emerging efforts combine tumor organoids with microfluidics to create more physiologically relevant models. By integrating mini-tumors or organoids on a chip, researchers better mimic tumor heterogeneity and ECM context while enabling drug response studies in a dynamic environment (Figure 1C,D) [52,53]. Collectively, these works provide proof-of-concept that tumor-on-chip platforms, which, by extension, include mechano-OoC systems, are both feasible and promising for cancer research. While many tumor-on-chip studies focus on reconstructing tissue architecture and establishing perfusion, integrating explicit mechanical cues (e.g., matrix stiffness gradients or cyclic compression) remains challenging. To address matrix mechanics, researchers employ tunable hydrogels or biomaterial scaffolds (such as collagen, fibrin, gelatin, or synthetic polymers) whose properties (stiffness, viscoelasticity, porosity, crosslinking) can be varied [54,55]. Embedding cancer cells or tumor organoids in such matrices can recreate the stiffer, desmoplastic ECM often seen in tumors [56,57]. At the same time, microfluidic platforms naturally allow for controlled flow and perfusion: by tuning channel geometry, flow rates, and matrix permeability, one can impose interstitial flow, generate fluid shear, and create gradients of nutrients or drugs [20,58]. This approach is well-established in physiological organ chips (e.g., vascular or renal models) and can be adapted to tumor chips to study transport phenomena [59].
Chip designs that include endothelialized vasculature alongside stromal and tumor compartments allow detailed modeling of blood-tumor interactions. For instance, chips incorporating endothelial-lined channels adjacent to tumor or stromal compartments can replicate angiogenesis, immune cell trafficking, and drug transport across the vasculature. Such vascularized multi-tissue chips have been demonstrated in other OoC contexts, suggesting that tumor-on-chip efforts can leverage similar strategies to mimic angiogenic remodeling and metastatic intravasation. Including multiple cell types on-chip further enables the study of complex immune and stromal interactions under flow [60,61]. So far, few high-impact studies have explicitly applied dynamic mechanical stress (cyclic strain, compression) to tumor-on-chip systems [62]. This reflects a critical challenge: to date, mechano-OoC platforms that subject tumour tissues to engineered cyclic deformation or solid stress are still relatively rare [63]. In summary, while OoC technologies are mature for replicating tissue architecture, perfusion, and cellular heterogeneity, truly mechanobiology-aware OoC systems, which incorporate controlled mechanical stress and dynamic stimuli, remain largely experimental [14,64]. We argue that overcoming these hurdles is essential to harness the full potential of mechano-OoC in cancer research.
Dynamic mechanical loading applied externally should be clearly distinguished from solid stress generated endogenously by tumor growth. Solid stress arises from volumetric expansion of proliferating tumor cells within a mechanically confining microenvironment and results in sustained, inward-directed compressive forces with biological effects distinct from transient external strain. In tumor-on-chip platforms, such growth-induced solid stress can be modeled and quantified by exploiting microstructural confinement, for example by embedding tumor spheroids within mechanically defined hydrogel chambers or microfabricated cavities [65]. As spheroids expand, deformation of the surrounding matrix or integrated elastic microstructures enables estimation of solid stress [66]. Moreover, chip-level physical confinement—through controlled chamber geometry and wall or matrix stiffness—can be used to mimic the inward pressure exerted by the tumor capsule in vivo, providing a complementary mechanical modality to cyclic strain–based mechano-OoC designs.

2.4. Challenges, Standardization, and Path Toward Reproducible Mechano-OoC

For mechano-OoC to become widely applicable in research and preclinical settings, several challenges must be addressed. First, design parameters and reporting must be standardized. In other words, materials, flow conditions, geometry, and cell composition should be documented in sufficient detail to allow reproducibility across laboratories [67,68]. Without consistent metadata and reporting standards, it is difficult to compare results or replicate findings. Thus, the field must adopt clear guidelines so that mechano-OoC experiments can be faithfully reproduced and interpreted.
Second, many mechano-OoC devices are technically complex and have low throughput. Incorporating features like vascularization, dynamic strain, and multiple compartments often yields intricate chips that are labor-intensive to fabricate and operate. This complexity limits scalability: for applications like drug screening, a balance between physiological relevance and throughput is needed, but such trade-offs remain a major bottleneck [69]. Similarly, increasing the number of cell types (e.g., adding stromal, immune, and endothelial cells) and biomechanical stimuli enhances biological realism, but this also further complicates device design and data analysis. In short, controlling complexity while maintaining experimental control is a persistent challenge [70,71].
Third, capturing the effects of mechanical cues requires suitable sensors and readout modalities. Many current tumor-on-chip studies rely on endpoint assays or simple microscopy, but to observe dynamic processes under mechanical stimulation, real-time and high-dimensional readouts are needed [72,73]. On-chip sensors for measuring variables like oxygen, pH, interstitial pressure, or cell-generated forces would greatly enhance the ability to monitor mechano-OoC experiments in situ. Finally, validation remains a hurdle: while OoC models can emulate many in vivo features, mechano-OoC systems have seldom been benchmarked against clinical data or patient-derived models. Without such validation, the translational relevance of mechano-OoC findings remains uncertain. For example, comparing invasion or drug response in the chip to animal or patient data is crucial to confirm that the mechanical modulations produce physiologically meaningful effects [74,75,76].
In summary, although the engineering toolbox largely exists and tumor-on-chip platforms have shown promise, truly standardized and validated mechano-OoC systems remain rare. High-impact publications that integrate controlled mechanical cues with robust validation are still scarce. We argue that addressing these issues, and in particular standardization, scalability, real-time sensing, and cross-validation, will be essential for mechano-OoC to fulfill its promise in cancer research. Challenges facing mechano-OoC, such as standardization and scalability, depend not only on the engineering optimization of platforms but also require matching targeted detection technologies and data interpretation systems. Thus, the following sections will focus on strategies for sensing, detection, and data standardization.

3. Sensing, Readout, and Data Considerations: Toward Mechano-OoC as a Research Standard

3.1. Lessons from General OoC for Assays and Readouts

Engineering a mechano-OoC is only half the battle; equally important is how we measure and analyze the biological responses. Lessons from general OoC development emphasize that readouts must be tailored for microfluidic systems. Conventional methods, such as microscopy (phase-contrast, fluorescence), live/dead assays, and standard immunostaining, can be applied. Still, work focusing on OoCs highlights the importance of compatibility with on-chip culture, such as optical access and non-destructive sampling. More advanced functional assays, which include barrier permeability tests, trans-endothelial electrical resistance, and tracer-based flow analysis, are recommended to probe dynamic behaviors [77,78,79]. In cancer models, these approaches should be combined with tumor-specific readouts (e.g., invasion/migration assays, proliferation markers, hypoxia sensors, drug response assays) to capture clinically relevant endpoints [80,81]. We also note that static endpoint measurements are often insufficient in mechanobiology contexts: they can miss how cells adapt over time to changing forces or remodel the ECM [82]. Therefore, real-time, longitudinal, and multimodal readouts are necessary to exploit mechano-OoC systems fully [83].

3.2. Emerging Readout Modalities Relevant for Mechano-OoC

New readout modalities are emerging for mechano-optical coherence tomography. Because chips are often made from transparent materials, live-cell imaging techniques (such as phase-contrast, fluorescence, and confocal microscopy) can be employed to track cell morphology, migration, and interactions under flow conditions. Indeed, several tumor-on-chip studies have used time-lapse microscopy to monitor tumor growth, organoid dynamics, and drug responses in perfused environments. Time-lapse microscopy can not only monitor tumor growth, but also quantify mechanotransduction-related phenotypes including the migration rate and nuclear deformation degree of tumor cells under mechanical stimulation [50]. Barrier integrity and permeability assays are particularly relevant for vascularized chips. In microfluidic devices with an endothelial barrier, one can measure transendothelial electrical resistance or track the diffusion of fluorescent tracers to quantify barrier function under flow [84,85]. These approaches, recommended in foundational OoC research, are valuable for modeling metastasis (intravasation/extravasation) and drug delivery across vessel walls [86].
Microfluidic perfusion itself offers a wealth of experimental possibilities (Figure 2A) [87]. By generating controlled gradients of nutrients, oxygen, or drugs across the chip, researchers can study how flow and ECM properties together influence drug penetration and efficacy [88,89,90]. For example, several tumor-on-chip systems have leveraged perfused microfluidic flows to perform drug screening under physiologically relevant transport conditions (Figure 2B) [91,92,93]. Mechano-OoC platforms can also integrate multiple cell types on-chip, and co-culturing tumor cells with stromal fibroblasts, endothelial cells, or immune cells in a perfused microenvironment allows studying complex processes like immune infiltration, stromal remodeling, angiogenesis, and therapy response under mechanical cues [94,95]. Although such sophisticated multi-cell chips are more common in non-cancer organ-chip models, the same design principles, such as multi-cell co-culture under perfused mechanical microenvironments, apply to tumor-on-chip systems [96,97].
Most tumor-on-chip platforms rely on pre-patterned, endothelial-lined microchannels to model perfusion and drug delivery; however, such architectures fail to recapitulate the structural irregularity and leakage characteristic of tumor angiogenesis. In contrast, self-assembled microvascular networks formed via endothelial sprouting more closely reproduce heterogeneous vessel diameters, disrupted junctions, and localized leakage, which are key contributors to elevated tumor interstitial fluid pressure [98]. Because vascular permeability directly governs interstitial pressure buildup and convective drug transport, tumor chips that lack physiologically relevant vascular leakage may substantially underestimate transport barriers and drug exposure gradients. Incorporating self-assembled vasculature therefore provides a more faithful mechanical and transport microenvironment for modeling tumor interstitial pressure and drug delivery than idealized pre-patterned channels. Finally, there is great potential to integrate on-chip sensors. For instance, embedded microsensors could continuously monitor oxygen tension, pH, interstitial pressure, matrix deformation, or cell-generated traction forces within the chip [99,100]. While mechano-OoC studies with such integrated sensors are still rare, developing these capabilities should be a priority in future work [101,102].

3.3. Sensor-Integrated and AI-Enhanced Readouts for Mechano-OoC

Recent progress has moved well beyond conventional imaging and trans-epithelial/endothelial electrical resistance (TEER), introducing sensor-integrated platforms that provide continuous, multi-dimensional monitoring of mechanical and biochemical signals inside Organ-on-Chip systems [103]. These technologies address a critical gap by enabling real-time mechanobiology analysis and improving translational relevance, particularly in dynamic microenvironments where cyclic strain, shear stress, and perfusion gradients shape cellular behavior. Conventional imaging and endpoint assays remain useful, but they cannot capture the complex temporal patterns of mechanotransduction [104,105,106]. To overcome this limitation, researchers are embedding advanced sensors directly into chip architectures and pairing them with AI-driven analytics, a shift that transforms Organ-on-Chip models into adaptive, high-content systems capable of delivering mechanistic insights with unprecedented precision [107].
One important advance is the creation of moisture-permeable, deformable circuits that remain electrically stable under mechanical strain and fluid exposure. Designs such as liquid-diode electronics enable the direct integration of sensors into microfluidic chips without compromising biocompatibility or flow performance in Figure 3 [108]. These embedded systems can continuously monitor pressure, strain, and barrier integrity during physiologic perfusion, supporting long-term studies of tissue mechanics and drug transport. Incorporating these electronics into chip substrates enables real-time mapping of stress distribution, ECM deformation, and interstitial pressure, reducing the need for invasive probes. This approach also supports multiplexed sensing, allowing oxygen tension, pH, and TEER to be tracked alongside mechanical cues [109]. The combination of soft electronics and microfluidics offers a practical route toward clinically relevant Organ-on-Chip platforms, bridging the gap between early prototypes and regulatory-grade models.

3.4. Liquid Metal Flexible Sensors Empower Microfluidic Models

Wearable sensors such as pressure devices and pulse monitors provide exceptional sensitivity to subtle mechanical changes (Figure 4A) [110,111]. Originally developed for cardiovascular monitoring, these sensors detect micro-scale pressure fluctuations and dynamic strain patterns; capabilities that translate effectively to mechano-OoC systems. When adapted for chip integration, they enable precise measurement of interstitial pressure, perfusion dynamics, and matrix deformation under controlled mechanical stimuli. Coupling these sensors with machine learning algorithms enhances interpretability, allowing automated classification of mechanical signatures linked to pathological states such as tumor stiffening or endothelial barrier failure [112]. In Organ-on-Chip contexts, this integration enables the real-time prediction of tissue responses under cyclic strain or shear, supporting adaptive drug dosing and mechanobiology-informed therapy design. AI-assisted sensing transforms OoC platforms from static observational models into dynamic, predictive systems.
Beyond sensor integration, recent efforts have shifted toward microfluidic 3D disease models, such as bladder cancer chips that incorporate bacterial-targeted aggregation-induced emission (AIE) photosensitizers for combined photodynamic and chemotherapeutic treatment [113]. This approach highlights how mechanical transport and barrier properties can significantly influence therapeutic outcomes, since drug penetration and light delivery depend on perfusion rates and ECM stiffness [114,115]. Adding sensors to monitor oxygen gradients, pH changes, and interstitial pressure enables these platforms to adjust treatment conditions dynamically, improving precision and efficacy. Introducing microbial components adds further complexity, both mechanical and biochemical, mimicking infection-driven inflammation, which is increasingly recognized as a factor in cancer progression [116]. These hybrid systems represent the next generation of mechano-OoC platforms: multimodal, sensor-rich, and clinically relevant, capable of revealing how mechanical and microbial cues interact to shape tumor biology and drug response. The multimodal Mechano-OoC platforms that integrate photomedicine and microbial cues generate multidimensional data, which require unified standardized procedures in particular to ensure reproducibility. Therefore, there is an urgent need to establish clear data reporting standards.

3.5. Data Standardization, Metadata, and Path Toward Reproducibility & Translation

To promote the wide adoption of mechano-OoC, researchers must accompany data generation with careful metadata reporting, standardization, and data-sharing (Figure 4B) [117,118,119]. Researchers should report all mechanical parameters in detail: ECM composition and stiffness, viscoelasticity, matrix architecture (fiber density, orientation), gel crosslinking, flow rates and shear stresses, pressure gradients, imposed deformation schedules (strain amplitude/frequency, compression cycles), device geometry and confinement dimensions, cell types and densities, culture conditions, and so on [102,120,121,122]. Likewise, for each readout, one should document the imaging modality (magnification, resolution, and channels), time-lapse schedule, tracer concentrations, assay protocols, and data analysis method [123,124,125].
We also advocate for sharing raw data and using standard file formats (e.g., raw imaging data, metadata in JSON/CSV, plate maps, time logs) [126]. This practice will enhance reproducibility, allow cross-study comparisons, and eventually enable pooled data sets for computational and machine-learning analyses [127]. Importantly, mechano-OoC results should be validated against in vivo or clinical data whenever possible. Comparing outcomes, such as invasion, metastasis, drug response, vascular permeability, or immune infiltration, between chip models and animal or patient-derived models is key to demonstrating translational relevance [128]. Only with such rigorous standards and validation can mechano-OoC approaches fulfill their potential as predictive preclinical platforms.
Figure 4. Mechano-OoC relies on two core components: sensor-integrated AI-enhanced readouts, and data standardization, metadata, along with the path to reproducibility and translation. (A) The liquid metal flexible sensor, fabricated via soft lithography, exhibits stable performance in reciprocating bending tests with a 6 mm bending radius, facilitates accurate joint bending angle detection, and realizes wireless Bluetooth car control after integration into a wearable glove [111]. (B) The tumor-on-a-chip system overview and mathematical model conditions for spheroids and on-chip scenarios involve a microfluidic system with side channels delivering DARPins to a tumor channel, a COMSOL-simulated spheroid model and a 3D on-chip geometry in COMSOL (COMSOL v6.3) accounting for distinct diffusion coefficients and void fractions in the tumor compartment and side channels [119].
Figure 4. Mechano-OoC relies on two core components: sensor-integrated AI-enhanced readouts, and data standardization, metadata, along with the path to reproducibility and translation. (A) The liquid metal flexible sensor, fabricated via soft lithography, exhibits stable performance in reciprocating bending tests with a 6 mm bending radius, facilitates accurate joint bending angle detection, and realizes wireless Bluetooth car control after integration into a wearable glove [111]. (B) The tumor-on-a-chip system overview and mathematical model conditions for spheroids and on-chip scenarios involve a microfluidic system with side channels delivering DARPins to a tumor channel, a COMSOL-simulated spheroid model and a 3D on-chip geometry in COMSOL (COMSOL v6.3) accounting for distinct diffusion coefficients and void fractions in the tumor compartment and side channels [119].
Ijms 27 01330 g004

4. Artificial Intelligence in Organ-on-Chip: From Image Analysis to Predictive Modeling

4.1. AI-Driven Image Analysis for Non-Destructive Evaluation in OoC Systems

Image-based analysis is the most mature and widely used entry point of artificial intelligence (AI) in Organ-on-Chip research. OoC platforms are inherently compatible with live imaging techniques, including bright-field, fluorescence, and confocal microscopy, enabling continuous monitoring of biological processes under physiologically relevant conditions [129,130,131]. However, extracting quantitative and reproducible information from such live imaging data remains challenging when relying on manual or conventional image-processing methods.
Furthermore, deep learning models, particularly convolutional neural networks (CNNs) are increasingly employed to analyze live imaging data generated by OoC systems [132]. These models enable automated evaluation of cellular morphology, tissue organization, and dynamic structural changes without the need for invasive measurements or destructive endpoint assays [131]. By learning complex spatial features directly from raw images, CNN-based approaches can accurately segment cells and tissues, quantify morphological heterogeneity, and track structural evolution over time [133,134]. Importantly, AI-driven image analysis supports the assessment of functional properties such as barrier integrity and tissue maturation in a non-destructive manner [135]. For example, CNNs can infer barrier formation, disruption, or remodeling by analyzing spatiotemporal patterns in fluorescence or phase-contrast images, reducing reliance on invasive permeability assays or labeling-based measurements. For the mechanical simulation scenarios of mechano-OoC, AI can automatically identify the correlation patterns between matrix stiffness and cell traction force through label-free phase contrast microscopy images, and quantify the efficiency of mechanotransduction without relying on fluorescent labeling. In addition, CNNs can not only automatically segment tumor cells, but also quantify key mechanotransduction phenotypes such as cell morphological heterogeneity induced by mechanical stimuli such as matrix stiffness gradients, including long axis to short axis ratio, nuclear deformation degree, thus avoiding the subjectivity of traditional manual analysis. This capability is particularly valuable for long-term OoC experiments, where repeated destructive measurements would otherwise compromise system stability.
Overall, the application of deep learning-based image analysis enhances the ability of OoC platforms to serve as continuous, non-invasive probes of living biological systems [132,133]. By enabling real-time, label-free, and longitudinal quantification of cellular and tissue-level features, AI tools significantly expand the experimental scope of OoC research and lay the foundation for subsequent multimodal integration and predictive modeling. This technology, AI image analysis, provides mechano-OoC with a solution for high-throughput quantification of mechanical phenotypes, resolving the key challenge that traditional methods are unable to monitor the temporal relationship between dynamic mechanical stimuli and cellular responses in real time.

4.2. From Multimodal Feature Extraction to Predictive Modeling in OoC Systems

The core requirement of mechano-OoC is to uncover the complex correlations among mechanical cues, biochemical signals, and cellular behaviors. AI multimodal predictive modeling is exactly able to capture such nonlinear interactions, and its core value lies in transforming mechanical mechanisms that cannot be directly measured into predictable clinical outcomes. While AI-driven image analysis provides a powerful and non-destructive means to quantify biological phenotypes in OoC systems, predictive modeling in this context should not rely solely on image-derived information. Instead, robust prediction typically emerges from the integration of multiple feature types, including image-based features, manually engineered descriptors, and system-level experimental parameters [136,137]. In OoC platforms, deep learning-based image analysis yields high-dimensional representations of cellular morphology, tissue organization, growth dynamics, and spatiotemporal behavior [136]. These image-derived features capture rich phenotypic information and often serve as primary inputs for predictive models. However, many biologically and experimentally relevant variables—such as flow rates, drug concentrations, culture duration, cell seeding density, matrix composition, and barrier permeability measurements—are not fully encoded in imaging data and must be incorporated through manual or conventional quantitative analyses [138,139].
Accordingly, recent AI-enabled OoC studies increasingly adopt hybrid feature spaces that combine automatically extracted image features with manually defined descriptors and experimental metadata. Machine learning models trained on such multimodal inputs can more accurately predict biological outcomes, including tumor growth trajectories, barrier integrity changes, and therapeutic responses, than models relying on imaging data alone. In cancer-on-chip systems, for example, predictive performance is substantially improved when image-based phenotypic features are integrated with treatment conditions and functional assay results, such as viability metrics or permeability measurements [140]. Importantly, manual feature engineering remains valuable in OoC predictive modeling, particularly for capturing domain-specific knowledge. Features derived from classical analyses—such as invasion depth, growth rate constants, dose–response parameters, or transport coefficients—provide interpretable and biologically grounded inputs that complement deep learning-based representations [136]. When combined with image-derived features, these manually curated parameters enhance both model robustness and interpretability [138].
Beyond feature integration, machine learning frameworks are well suited to capture nonlinear interactions among heterogeneous inputs. Supervised learning models can associate early-stage multimodal features with downstream outcomes, enabling prediction of long-term system behavior or treatment efficacy before experimental endpoints are reached. Such predictive capabilities are especially valuable for long-term OoC experiments, where early intervention or adaptive experimental design can reduce cost and experimental burden [136]. Emerging approaches further explore hybrid predictive strategies that integrate data-driven learning with mechanistic or physics-informed constraints. By embedding prior biological knowledge or transport and growth models into machine learning pipelines, these approaches aim to improve generalizability across OoC designs and experimental conditions while maintaining predictive accuracy. In the predictive modeling of Mechano-OoC, features associated with dynamic mechanical changes should be incorporated as key elements, including the evolution curve of matrix stiffness during tumor growth and the pulse frequency of shear force. Such features can notably enhance the prediction accuracy of the model regarding tumor metastasis and drug penetration depth. For example, when stiffness gradient ranging from 1 to 10 kPa and drug IC50 value are adopted as combined inputs, the prediction error of the AI model for chemotherapy efficacy can be reduced by more than 30% [141]. Although still at an early stage, such frameworks represent an important step toward transferable and clinically relevant OoC-based prediction.
Overall, predictive modeling in OoC systems is a multimodal process, integrating AI-derived image features, manually analyzed experimental variables, and system-level metadata [140]. This holistic approach enables OoC platforms to transition from descriptive microphysiological models toward predictive and decision-support tools, with broad implications for drug development, disease modeling, and precision medicine.

4.3. Challenges and Future Perspectives

Despite rapid progress, important challenges remain. AI performance depends critically on data quality, consistency, and annotation, reinforcing the need for standardized OoC protocols and comprehensive metadata reporting [137,141]. In addition, most AI models are developed for specific chip designs or experimental contexts, limiting their transferability across laboratories and platforms. Nonetheless, a consensus has emerged across extant research that artificial intelligence will constitute an indispensable, intrinsic component of Organ-on-Chip research workflows. As OoC platforms advance in maturity, and as the scale and heterogeneity of corresponding datasets expand, AI-driven image analysis, multimodal data integration, and predictive computational modeling are poised to assume increasingly central functional roles. Ultimately, the synergistic convergence of AI and OoC technologies carries considerable promise for the development of predictive, patient-centric in vitro models, which establish a critical translational interface between experimental biology and clinical decision-making. The integration of AI and Mechano-OoC, despite challenges such as data quality and model transferability, has demonstrated the potential for full-chain empowerment, covering automatic identification of mechanical phenotypes, integration of multimodal data, and prediction of therapeutic efficacy. This, together with the platform engineering and sensing technologies mentioned earlier, jointly advances the clinical translation of mechano-OoC.

5. Discussion

Pairing time-lapse microscopy with embedded sensors and machine learning is transforming how data is captured in Organ-on-Chip systems. Instead of relying on manual observation, these integrated setups can automatically quantify cell migration, traction forces, and barrier dynamics under cyclic strain and shear. Advanced algorithms now merge imaging streams with sensor outputs to build composite maps of mechanical stress and biochemical flux across the chip. This combined approach accelerates interpretation and enables adaptive control of experimental conditions. For instance, AI can detect early signs of endothelial barrier compromise and adjust perfusion rates or drug dosing in real time; functionality that mirrors clinical feedback systems. Such capabilities move Organ-on-Chip platforms beyond static observation toward responsive experimental ecosystems, where mechanical and biochemical signals actively guide decisions. By integrating soft electronics, microfluidics, and AI-driven analytics, these sensor-rich strategies provide a foundation for precision medicine and mechanobiology-informed drug development. Real-time fusion of AI image analysis and embedded sensor data enables dynamic correlation analysis between mechanical stimuli such as cyclic compression and cellular phenotypes such as EMT marker expression. For example, by monitoring the degree of nuclear deformation, AI can predict matrix stiffness-induced drug resistance in real time, providing a reference for the adjustment of clinical individualized treatment regimens.

6. Conclusions

In summary, we have argued that the mechanical landscape of tumors, which encompasses factors from matrix stiffness and solid stress to fluid flow and confinement, profoundly influences cancer progression and therapy response. Conventional models often neglect these forces, whereas Organ-on-Chip systems offer the means to recapitulate tissue architecture, perfusion, and controlled mechanics. We emphasize that tumor-on-chip efforts must evolve to integrate mechanobiology explicitly: incorporating tunable ECM, dynamic loading, live sensing, and complex multicellular culture. Achieving this vision will require standardized platform designs and data reporting, scalable device architectures, and rigorous validation against in vivo data. By addressing these challenges, mechano-OoC could emerge as a powerful tool for studying invasion, metastasis, and treatment efficacy under realistic biophysical conditions. The mechano-OoC platforms outlined here effectively overcome the shortcomings of 2D cultures that lack mechanical cues and animal models with large species differences, through the integration of modules including tunable ECM stiffness, dynamic mechanical loading, and multicellular co-culture. Meanwhile, standardized data reporting and clinical sample verification further boost their reliability when used to replace traditional models in drug screening and mechanism-based research. By integrating four modules, engineerable tunable mechanical microenvironment, multimodal sensing, data standardization, and AI-driven interpretation, mechano-OoC not only overcomes the mechanical simulation limitations of traditional models, but also achieves the integration of mechanism elucidation and therapeutic efficacy prediction, providing a novel tool for mechanobiology-guided precision cancer therapy. The incorporation of AI technology further resolves the problems of low efficiency and insufficient predictability in the interpretation of multi-dimensional mechano-OoC data. Combined with standardized design and clinical validation, this enables mechano-OoC to become an integrated platform for mechanical mechanism research, drug screening, and therapeutic efficacy prediction.

Author Contributions

Conceptualization, S.H.D.W., B.L.K. and X.Z.; software, L.W. and J.C.W.C.; writing—original draft preparation, L.W., J.C.W.C. and X.Z.; writing—review and editing, L.W., J.C.W.C. and B.L.K.; visualization, L.W., S.H.D.W. and B.L.K.; supervision, S.H.D.W. and B.L.K.; project administration, S.H.D.W.; funding acquisition, S.H.D.W. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by National Natural Science Foundation of China Young Scientists Fund (32501158), Start-up Fundings of Ocean University of China (862401013154 and 862401013155), Laboratory for Marine Drugs and Bioproducts Qingdao Marine Science and Technology Center (no.: LMDBCXRC202401 and LMDBCXRC202402), Taishan Scholar Youth Expert Program of Shandong Province (tsqn202306102 and tsqn202312105), and Shandong Provincial Overseas Excellent Young Scholar Program (2024HWYQ-042 and 2024HWYQ-043).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare that they have no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
Mechano-OoCMechano-Organ-on-Chip
TMETumor microenvironment
ECMExtracellular matrix
EMTEpithelial–mesenchymal transition
OoCOrgan-on-chip
PDMSPolydimethylsiloxane
PSPolystyrene
PMMAPolymethyl methacrylate
COC/COPCyclic olefin copolymer/polymer
GelMAGelatin methacrylate
LAPLithium phenyl-2,4,6-trimethylbenzoylphosphinate
PEGDAPoly(ethylene glycol) diacrylate
DLPDigital light processing
CADComputer-aided design
TEERTrans-epithelial/endothelial electrical resistance
AIEAggregation-induced emission
DARPinsDesigned ankyrin repeat proteins
COMSOLCOMSOL multiphysics
AIArtificial intelligence

References

  1. Marrella, A.; Fedi, A.; Varani, G.; Vaccari, I.; Fato, M.; Firpo, G.; Guida, P.; Aceto, N.; Scaglione, S. High blood flow shear stress values are associated with circulating tumor cells cluster disaggregation in a multi-channel microfluidic device. PLoS ONE 2021, 16, e0245536. [Google Scholar] [CrossRef]
  2. Yankaskas, C.L.; Bera, K.; Stoletov, K.; Serra, S.A.; Carrillo-Garcia, J.; Tuntithavornwat, S.; Mistriotis, P.; Lewis, J.D.; Valverde, M.A.; Konstantopoulos, K. The fluid shear stress sensor TRPM7 regulates tumor cell intravasation. Sci. Adv. 2021, 7, eabh3457. [Google Scholar] [CrossRef] [PubMed]
  3. Wong, S.H.D.; Yin, B.; Li, Z.; Yuan, W.; Zhang, Q.; Xie, X.; Tan, Y.; Wong, N.; Zhang, K.; Bian, L. Mechanical manipulation of cancer cell tumorigenicity via heat shock protein signaling. Sci. Adv. 2023, 9, eadg9593. [Google Scholar] [CrossRef] [PubMed]
  4. Aydin, H.B.; Ozcelikkale, A.; Acar, A. Exploiting Matrix Stiffness to Overcome Drug Resistance. ACS Biomater. Sci. Eng. 2024, 10, 4682–4700. [Google Scholar] [CrossRef]
  5. Zhou, H.; Wang, M.; Zhang, Y.; Su, Q.; Xie, Z.; Chen, X.; Yan, R.; Li, P.; Li, T.; Qin, X. Functions and clinical significance of mechanical tumor microenvironment: Cancer cell sensing, mechanobiology and metastasis. Cancer Commun. 2022, 42, 374–400. [Google Scholar] [CrossRef] [PubMed]
  6. Barbazan, J.; Perez-Gonzalez, C.; Gomez-Gonzalez, M.; Dedenon, M.; Richon, S.; Latorre, E.; Serra, M.; Mariani, P.; Descroix, S.; Sens, P.; et al. Cancer-associated fibroblasts actively compress cancer cells and modulate mechanotransduction. Nat. Commun. 2023, 14, 6966. [Google Scholar] [CrossRef]
  7. Wu, B.; Liu, D.A.; Guan, L.; Myint, P.K.; Chin, L.; Dang, H.; Xu, Y.; Ren, J.; Li, T.; Yu, Z.; et al. Stiff matrix induces exosome secretion to promote tumour growth. Nat. Cell Biol. 2023, 25, 415–424, Correction in Nat. Cell Biol. 2024, 26, 490–491. [Google Scholar] [CrossRef]
  8. Zamprogno, P.; Wuthrich, S.; Achenbach, S.; Thoma, G.; Stucki, J.D.; Hobi, N.; Schneider-Daum, N.; Lehr, C.M.; Huwer, H.; Geiser, T.; et al. Second-generation lung-on-a-chip with an array of stretchable alveoli made with a biological membrane. Commun. Biol. 2021, 4, 168. [Google Scholar] [CrossRef]
  9. Grebenyuk, S.; Abdel Fattah, A.R.; Kumar, M.; Toprakhisar, B.; Rustandi, G.; Vananroye, A.; Salmon, I.; Verfaillie, C.; Grillo, M.; Ranga, A. Large-scale perfused tissues via synthetic 3D soft microfluidics. Nat. Commun. 2023, 14, 193. [Google Scholar] [CrossRef]
  10. Quintard, C.; Tubbs, E.; Jonsson, G.; Jiao, J.; Wang, J.; Werschler, N.; Laporte, C.; Pitaval, A.; Bah, T.S.; Pomeranz, G.; et al. A microfluidic platform integrating functional vascularized organoids-on-chip. Nat. Commun. 2024, 15, 1452. [Google Scholar] [CrossRef]
  11. Fang, G.; Chen, Y.C.; Lu, H.; Jin, D. Advances in Spheroids and Organoids on a Chip. Adv. Funct. Mater. 2023, 33, 2215043. [Google Scholar] [CrossRef]
  12. Ge, J.Y.; Wang, Y.; Li, Q.L.; Liu, F.K.; Lei, Q.K.; Zheng, Y.W. Trends and challenges in organoid modeling and expansion with pluripotent stem cells and somatic tissue. PeerJ 2024, 12, e18422. [Google Scholar] [CrossRef]
  13. Jin, J.; Yoshimura, K.; Sewastjanow-Silva, M.; Song, S.; Ajani, J.A. Challenges and Prospects of Patient-Derived Xenografts for Cancer Research. Cancers 2023, 15, 4352. [Google Scholar] [CrossRef]
  14. Wang, H.; Ning, X.; Zhao, F.; Zhao, H.; Li, D. Human organoids-on-chips for biomedical research and applications. Theranostics 2024, 14, 788–818. [Google Scholar] [CrossRef]
  15. Abizanda-Campo, S.; Virumbrales-Munoz, M.; Humayun, M.; Marmol, I.; Beebe, D.J.; Ochoa, I.; Olivan, S.; Ayuso, J.M. Microphysiological systems for solid tumor immunotherapy: Opportunities and challenges. Microsyst. Nanoeng. 2023, 9, 154. [Google Scholar] [CrossRef]
  16. Peng, Y.; Lee, E. Microphysiological Systems for Cancer Immunotherapy Research and Development. Adv. Biol. 2024, 8, e2300077. [Google Scholar] [CrossRef] [PubMed]
  17. Zhao, Y.; Wang, E.Y.; Lai, F.B.L.; Cheung, K.; Radisic, M. Organs-on-a-chip: A union of tissue engineering and microfabrication. Trends Biotechnol. 2023, 41, 410–424. [Google Scholar] [CrossRef] [PubMed]
  18. Peel, S.; Jackman, M. Imaging microphysiological systems: A review. Am. J. Physiol.-Cell Physiol. 2021, 320, C669–C680. [Google Scholar] [CrossRef]
  19. Zhang, S.; Xu, G.; Wu, J.; Liu, X.; Fan, Y.; Chen, J.; Wallace, G.; Gu, Q. Microphysiological Constructs and Systems: Biofabrication Tactics, Biomimetic Evaluation Approaches, and Biomedical Applications. Small Methods 2024, 8, e2300685. [Google Scholar] [CrossRef]
  20. Skubal, M.; Larney, B.M.; Phung, N.B.; Desmaras, J.C.; Dozic, A.V.; Volpe, A.; Ogirala, A.; Machado, C.L.; Djibankov, J.; Ponomarev, V.; et al. Vascularized tumor on a microfluidic chip to study mechanisms promoting tumor neovascularization and vascular targeted therapies. Theranostics 2025, 15, 766–783. [Google Scholar] [CrossRef]
  21. Chakrabarty, S.; Quiros-Solano, W.F.; Kuijten, M.M.P.; Haspels, B.; Mallya, S.; Lo, C.S.Y.; Othman, A.; Silvestri, C.; van de Stolpe, A.; Gaio, N.; et al. A Microfluidic Cancer-on-Chip Platform Predicts Drug Response Using Organotypic Tumor Slice Culture. Cancer Res. 2022, 82, 510–520. [Google Scholar] [CrossRef]
  22. Monteiro, C.F.; Deus, I.A.; Silva, I.B.; Duarte, I.F.; Custódio, C.A.; Mano, J.F. Tumor-On-A-Chip Model Incorporating Human-Based Hydrogels for Easy Assessment of Metastatic Tumor Inter-Heterogeneity. Adv. Funct. Mater. 2024, 34, 2315940. [Google Scholar] [CrossRef]
  23. Liang, L.; Song, X.; Zhao, H.; Lim, C.T. Insights into the mechanobiology of cancer metastasis via microfluidic technologies. APL Bioeng. 2024, 8, 021506. [Google Scholar] [CrossRef]
  24. Gil, J.F.; Moura, C.S.; Silverio, V.; Goncalves, G.; Santos, H.A. Cancer Models on Chip: Paving the Way to Large-Scale Trial Applications. Adv. Mater. 2023, 35, e2300692. [Google Scholar] [CrossRef]
  25. Souza, I.F.; Vieira, J.P.J.; Bonifácio, E.D.; Avelar Freitas, B.A.d.; Torres, L.A.G. The Microenvironment of Solid Tumors: Components and Current Challenges of Tumor-on-a-Chip Models. Tissue Eng. Part B Rev. 2025, 31, 266–283. [Google Scholar] [CrossRef]
  26. Mai, Z.; Lin, Y.; Lin, P.; Zhao, X.; Cui, L. Modulating extracellular matrix stiffness: A strategic approach to boost cancer immunotherapy. Cell Death Dis. 2024, 15, 307. [Google Scholar] [CrossRef]
  27. Denais, C.M.; Gilbert, R.M.; Isermann, P.; McGregor, A.L.; te Lindert, M.; Weigelin, B.; Davidson, P.M.; Friedl, P.; Wolf, K.; Lammerding, J. Nuclear envelope rupture and repair during cancer cell migration. Science 2016, 352, 353–358. [Google Scholar] [CrossRef]
  28. Nguyen, H.-T.; Rissanen, S.-L.; Peltokangas, M.; Laakkonen, T.; Kettunen, J.; Barthod, L.; Sivakumar, R.; Palojärvi, A.; Junttila, P.; Talvitie, J.; et al. Highly scalable and standardized organ-on-chip platform with TEER for biological barrier modeling. Tissue Barriers 2024, 12, 2315702. [Google Scholar] [CrossRef]
  29. Chaudhuri, O.; Gu, L.; Klumpers, D.; Darnell, M.; Bencherif, S.A.; Weaver, J.C.; Huebsch, N.; Lee, H.P.; Lippens, E.; Duda, G.N.; et al. Hydrogels with tunable stress relaxation regulate stem cell fate and activity. Nat. Mater. 2016, 15, 326–334. [Google Scholar] [CrossRef]
  30. Zhang, M.; Zhang, B. Extracellular matrix stiffness: Mechanisms in tumor progression and therapeutic potential in cancer. Exp. Hematol. Oncol. 2025, 14, 54. [Google Scholar] [CrossRef]
  31. Zhang, S.; Grifno, G.; Passaro, R.; Regan, K.; Zheng, S.; Hadzipasic, M.; Banerji, R.; O’Connor, L.; Chu, V.; Kim, S.Y.; et al. Intravital measurements of solid stresses in tumours reveal length-scale and microenvironmentally dependent force transmission. Nat. Biomed. Eng. 2023, 7, 1473–1492. [Google Scholar] [CrossRef]
  32. Stylianopoulos, T.; Martin, J.D.; Chauhan, V.P.; Jain, S.R.; Diop-Frimpong, B.; Bardeesy, N.; Smith, B.L.; Ferrone, C.R.; Hornicek, F.J.; Boucher, Y.; et al. Causes, consequences, and remedies for growth-induced solid stress in murine and human tumors. Proc. Natl. Acad. Sci. USA 2012, 109, 15101–15108. [Google Scholar] [CrossRef]
  33. Joshi, I.M.; Mansouri, M.; Ahmed, A.; De Silva, D.; Simon, R.A.; Esmaili, P.; Desa, D.E.; Elias, T.M.; Brown, E.B.; Abhyankar, V.V. Microengineering 3D Collagen Matrices with Tumor-Mimetic Gradients in Fiber Alignment. Adv. Funct. Mater. 2023, 34, 2308071. [Google Scholar] [CrossRef]
  34. Kamaras, C.; Frank, D.; Wang, H.; Drepper, F.; Huesgen, P.F.; Grosse, R. Nuclear rupture in confined cell migration triggers nuclear actin polymerization to limit chromatin leakage. EMBO J. 2025, 44, 6112–6136. [Google Scholar] [CrossRef]
  35. Hopkins, E.; Valois, E.; Stull, A.; Le, K.; Pitenis, A.A.; Wilson, M.Z. An Optogenetic Platform to Dynamically Control the Stiffness of Collagen Hydrogels. ACS Biomater. Sci. Eng. 2021, 7, 408–414. [Google Scholar] [CrossRef]
  36. Smits, J.; van der Pol, A.; Goumans, M.J.; Bouten, C.V.C.; Jorba, I. GelMA hydrogel dual photo-crosslinking to dynamically modulate ECM stiffness. Front. Bioeng. Biotechnol. 2024, 12, 1363525. [Google Scholar] [CrossRef]
  37. Prakash, J.; Shaked, Y. The Interplay between Extracellular Matrix Remodeling and Cancer Therapeutics. Cancer Discov. 2024, 14, 1375–1388. [Google Scholar] [CrossRef]
  38. Kutluk, H.; Bastounis, E.E.; Constantinou, I. Integration of Extracellular Matrices into Organ-on-Chip Systems. Adv. Healthc. Mater. 2023, 12, e2203256. [Google Scholar] [CrossRef]
  39. Zhou, Z.; Vessella, T.; Wang, P.; Cui, F.; Wen, Q.; Zhou, H.S. Mechanical cues in tumor microenvironment on chip. Biosens. Bioelectron. X 2023, 14, 100376. [Google Scholar] [CrossRef]
  40. Huh, D.; Matthews, B.D.; Mammoto, A.; Montoya-Zavala, M.; Hsin, H.Y.; Ingber, D.E. Reconstituting organ-level lung functions on a chip. Science 2010, 328, 1662–1668. [Google Scholar] [CrossRef]
  41. Polacheck, W.J.; Charest, J.L.; Kamm, R.D. Interstitial flow influences direction of tumor cell migration through competing mechanisms. Proc. Natl. Acad. Sci USA 2011, 108, 11115–11120. [Google Scholar] [CrossRef]
  42. Stowers, R.S.; Allen, S.C.; Suggs, L.J. Dynamic phototuning of 3D hydrogel stiffness. Proc. Natl. Acad. Sci USA 2015, 112, 1953–1958. [Google Scholar] [CrossRef]
  43. Farhang Doost, N.; Srivastava, S.K. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. Biosensors 2024, 14, 225. [Google Scholar] [CrossRef]
  44. Iakovlev, A.P.; Erofeev, A.S.; Gorelkin, P.V. Novel Pumping Methods for Microfluidic Devices: A Comprehensive Review. Biosensors 2022, 12, 956. [Google Scholar] [CrossRef]
  45. Ferrari, D.; Sengupta, A.; Heo, L.; Petho, L.; Michler, J.; Geiser, T.; de Jesus Perez, V.A.; Kuebler, W.M.; Zeinali, S.; Guenat, O.T. Effects of biomechanical and biochemical stimuli on angio- and vasculogenesis in a complex microvasculature-on-chip. iScience 2023, 26, 106198. [Google Scholar] [CrossRef]
  46. Toepke, M.W.; Beebe, D.J. PDMS absorption of small molecules and consequences in microfluidic applications. Lab Chip 2006, 6, 1484–1486. [Google Scholar] [CrossRef]
  47. Campbell, S.B.; Wu, Q.; Yazbeck, J.; Liu, C.; Okhovatian, S.; Radisic, M. Beyond Polydimethylsiloxane: Alternative Materials for Fabrication of Organ-on-a-Chip Devices and Microphysiological Systems. ACS Biomater. Sci. Eng. 2021, 7, 2880–2899. [Google Scholar] [CrossRef]
  48. Sanchez-Salazar, M.G.; Crespo-Lopez Oliver, R.; Ramos-Meizoso, S.; Jerezano-Flores, V.S.; Gallegos-Martinez, S.; Bolivar-Monsalve, E.J.; Ceballos-Gonzalez, C.F.; Trujillo-de Santiago, G.; Alvarez, M.M. 3D-Printed Tumor-on-Chip for the Culture of Colorectal Cancer Microspheres: Mass Transport Characterization and Anti-Cancer Drug Assays. Bioengineering 2023, 10, 554. [Google Scholar] [CrossRef]
  49. Wang, L.; Tong, L.; Xiong, Z.; Chen, Y.; Zhang, P.; Gao, Y.; Liu, J.; Yang, L.; Huang, C.; Ye, G.; et al. Ferroptosis-inducing nanomedicine and targeted short peptide for synergistic treatment of hepatocellular carcinoma. J. Nanobiotechnol. 2024, 22, 533. [Google Scholar] [CrossRef]
  50. Li, C.; Holman, J.B.; Shi, Z.; Qiu, B.; Ding, W. On-chip modeling of tumor evolution: Advances, challenges and opportunities. Mater. Today Bio. 2023, 21, 100724. [Google Scholar] [CrossRef]
  51. Pinho, D.; Santos, D.; Vila, A.; Carvalho, S. Establishment of Colorectal Cancer Organoids in Microfluidic-Based System. Micromachines 2021, 12, 497. [Google Scholar] [CrossRef]
  52. Hwangbo, H.; Chae, S.; Kim, W.; Jo, S.; Kim, G.H. Tumor-on-a-chip models combined with mini-tissues or organoids for engineering tumor tissues. Theranostics 2024, 14, 33–55. [Google Scholar] [CrossRef]
  53. Amereh, M.; Seyfoori, A.; Dallinger, B.; Azimzadeh, M.; Stefanek, E.; Akbari, M. 3D-Printed Tumor-on-a-Chip Model for Investigating the Effect of Matrix Stiffness on Glioblastoma Tumor Invasion. Biomimetics 2023, 8, 421. [Google Scholar] [CrossRef]
  54. Wei, Z.; Lei, M.; Wang, Y.; Xie, Y.; Xie, X.; Lan, D.; Jia, Y.; Liu, J.; Ma, Y.; Cheng, B.; et al. Hydrogels with tunable mechanical plasticity regulate endothelial cell outgrowth in vasculogenesis and angiogenesis. Nat. Commun. 2023, 14, 8307, Correction in Nat. Commun. 2024, 15, 3274. [Google Scholar] [CrossRef]
  55. Sievers, J.; Mahajan, V.; Welzel, P.B.; Werner, C.; Taubenberger, A. Precision Hydrogels for the Study of Cancer Cell Mechanobiology. Adv. Healthc. Mater. 2023, 12, e2202514. [Google Scholar] [CrossRef]
  56. Maller, O.; Drain, A.P.; Barrett, A.S.; Borgquist, S.; Ruffell, B.; Zakharevich, I.; Pham, T.T.; Gruosso, T.; Kuasne, H.; Lakins, J.N.; et al. Tumour-associated macrophages drive stromal cell-dependent collagen crosslinking and stiffening to promote breast cancer aggression. Nat. Mater. 2021, 20, 548–559. [Google Scholar] [CrossRef]
  57. Liu, Y.; Okesola, B.O.; Osuna de la Peña, D.; Li, W.; Lin, M.L.; Trabulo, S.; Tatari, M.; Lawlor, R.T.; Scarpa, A.; Wang, W.; et al. A Self-Assembled 3D Model Demonstrates How Stiffness Educates Tumor Cell Phenotypes and Therapy Resistance in Pancreatic Cancer. Adv. Healthc. Mater. 2024, 13, 2301941. [Google Scholar] [CrossRef]
  58. de Roode, K.E.; Hashemi, K.; Verdurmen, W.P.R.; Brock, R. Tumor-On-A-Chip Models for Predicting In Vivo Nanoparticle Behavior. Small 2024, 20, 2402311. [Google Scholar] [CrossRef]
  59. Shaji, M.; Tamada, A.; Fujimoto, K.; Muguruma, K.; Karsten, S.L.; Yokokawa, R. Deciphering potential vascularization factors of on-chip co-cultured hiPSC-derived cerebral organoids. Lab Chip 2024, 24, 680–696. [Google Scholar] [CrossRef]
  60. Zhao, Y.; Wu, Y.; Islam, K.; Paul, R.; Zhou, Y.; Qin, X.; Li, Q.; Liu, Y. Microphysiologically Engineered Vessel-Tumor Model to Investigate Vascular Transport Dynamics of Immune Cells. ACS Appl. Mater. Interfaces 2024, 16, 22839–22849. [Google Scholar] [CrossRef]
  61. Wan, Z.; Floryan, M.A.; Coughlin, M.F.; Zhang, S.; Zhong, A.X.; Shelton, S.E.; Wang, X.; Xu, C.; Barbie, D.A.; Kamm, R.D. New Strategy for Promoting Vascularization in Tumor Spheroids in a Microfluidic Assay. Adv. Healthc. Mater. 2023, 12, 2201784. [Google Scholar] [CrossRef]
  62. Riveiro Rodríguez, A.; Onal, S.; Alkaisi, M.M.; Nock, V. Application of sequential cyclic compression on cancer cells in a flexible microdevice. PLoS ONE 2023, 18, e0279896. [Google Scholar]
  63. Mary, G.; Malgras, B.; Perez, J.E.; Nagle, I.; Luciani, N.; Pimpie, C.; Asnacios, A.; Pocard, M.; Reffay, M.; Wilhelm, C. Magnetic Compression of Tumor Spheroids Increases Cell Proliferation In Vitro and Cancer Progression In Vivo. Cancers 2022, 14, 366. [Google Scholar] [CrossRef]
  64. Cao, T.; Xu, P.; Yang, C.; Chen, Y.; Wang, Y.; Zhang, J.; Ye, F. Design strategy primer for organ-on-chips. Biomater. Transl. 2025, 6, 250–264. [Google Scholar]
  65. Stylianopoulos, T.; Jain, R.K. Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc. Natl. Acad. Sci. USA 2013, 110, 18632–18637. [Google Scholar] [CrossRef]
  66. Nia, H.T.; Liu, H.; Seano, G.; Datta, M.; Jones, D.; Rahbari, N.; Incio, J.; Chauhan, V.P.; Jung, K.; Martin, J.D.; et al. Solid stress and elastic energy as measures of tumour mechanopathology. Nat. Biomed. Eng. 2016, 1, 0004. [Google Scholar] [CrossRef]
  67. Mohapatra, R.; Leist, M.; von Aulock, S.; Hartung, T. Guidance for Good In Vitro Reporting Standards (GIVReSt)—A draft for stakeholder discussion and background documentation. ALTEX 2025, 42, 376–396. [Google Scholar]
  68. Pamies, D.; Ekert, J.; Zurich, M.G.; Frey, O.; Werner, S.; Piergiovanni, M.; Freedman, B.S.; Keong Teo, A.K.; Erfurth, H.; Reyes, D.R.; et al. Recommendations on fit-for-purpose criteria to establish quality management for microphysiological systems and for monitoring their reproducibility. Stem Cell Rep. 2024, 19, 604–617. [Google Scholar] [CrossRef]
  69. Liu, X.; Fang, J.; Huang, S.; Wu, X.; Xie, X.; Wang, J.; Liu, F.; Zhang, M.; Peng, Z.; Hu, N. Tumor-on-a-chip: From bioinspired design to biomedical application. Microsyst. Nanoeng. 2021, 7, 50. [Google Scholar]
  70. Cook, S.R.; Ball, A.G.; Mohammad, A.; Pompano, R.R. A 3D-printed multi-compartment organ-on-chip platform with a tubing-free pump models communication with the lymph node. Lab Chip 2025, 25, 155–174. [Google Scholar] [CrossRef]
  71. Vuorenpaa, H.; Bjorninen, M.; Valimaki, H.; Ahola, A.; Kroon, M.; Honkamaki, L.; Koivumaki, J.T.; Pekkanen-Mattila, M. Building blocks of microphysiological system to model physiology and pathophysiology of human heart. Front. Physiol. 2023, 14, 1213959. [Google Scholar] [CrossRef]
  72. Liu, Y.; Tian, Y.; Lin, C.; Miao, J.; Yu, X. A monolithically integrated microcantilever biosensor based on partially depleted SOI CMOS technology. Microsyst. Nanoeng. 2023, 9, 60. [Google Scholar] [CrossRef]
  73. Mou, L.; Mandal, K.; Mecwan, M.M.; Hernandez, A.L.; Maity, S.; Sharma, S.; Herculano, R.D.; Kawakita, S.; Jucaud, V.; Dokmeci, M.R.; et al. Integrated biosensors for monitoring microphysiological systems. Lab Chip 2022, 22, 3801–3816, Correction in Lab Chip 2022, 22, 3801–3816. [Google Scholar] [CrossRef]
  74. Sachs, N.; de Ligt, J.; Kopper, O.; Gogola, E.; Bounova, G.; Weeber, F.; Balgobind, A.V.; Wind, K.; Gracanin, A.; Begthel, H.; et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell 2018, 172, 373–386 e10. [Google Scholar] [CrossRef] [PubMed]
  75. Pauli, C.; Hopkins, B.D.; Prandi, D.; Shaw, R.; Fedrizzi, T.; Sboner, A.; Sailer, V.; Augello, M.; Puca, L.; Rosati, R.; et al. Personalized In Vitro and In Vivo Cancer Models to Guide Precision Medicine. Cancer Discov. 2017, 7, 462–477. [Google Scholar] [CrossRef]
  76. Vlachogiannis, G.; Hedayat, S.; Vatsiou, A.; Jamin, Y.; Fernandez-Mateos, J.; Khan, K.; Lampis, A.; Eason, K.; Huntingford, I.; Burke, R.; et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science 2018, 359, 920–926. [Google Scholar] [CrossRef]
  77. Aydogmus, H.; Hu, M.; Ivancevic, L.; Frimat, J.P.; van den Maagdenberg, A.; Sarro, P.M.; Mastrangeli, M. An organ-on-chip device with integrated charge sensors and recording microelectrodes. Sci. Rep. 2023, 13, 8062. [Google Scholar] [CrossRef]
  78. Bossink, E.; Zakharova, M.; de Bruijn, D.S.; Odijk, M.; Segerink, L.I. Measuring barrier function in organ-on-chips with cleanroom-free integration of multiplexable electrodes. Lab Chip 2021, 21, 2040–2049. [Google Scholar] [CrossRef] [PubMed]
  79. Dornhof, J.; Kieninger, J.; Muralidharan, H.; Maurer, J.; Urban, G.A.; Weltin, A. Microfluidic organ-on-chip system for multi-analyte monitoring of metabolites in 3D cell cultures. Lab Chip 2022, 22, 225–239. [Google Scholar] [CrossRef]
  80. Wang, L.; Wang, H.; Yang, C.; Wu, Y.; Lei, G.; Yu, Y.; Gao, Y.; Du, J.; Tong, X.; Zhou, F.; et al. Investigating CENPW as a Novel Biomarker Correlated With the Development and Poor Prognosis of Breast Carcinoma. Front. Genet. 2022, 13, 900111. [Google Scholar] [CrossRef] [PubMed]
  81. Zhou, C.; Wang, L.; Hu, W.; Tang, L.; Zhang, P.; Gao, Y.; Du, J.; Li, Y.; Wang, Y. CDC25C is a prognostic biomarker and correlated with mitochondrial homeostasis in pancreatic adenocarcinoma. Bioengineered 2022, 13, 13089–13107. [Google Scholar] [CrossRef] [PubMed]
  82. Stucki, A.O.; Stucki, J.D.; Hall, S.R.; Felder, M.; Mermoud, Y.; Schmid, R.A.; Geiser, T.; Guenat, O.T. A lung-on-a-chip array with an integrated bio-inspired respiration mechanism. Lab Chip 2015, 15, 1302–1310. [Google Scholar] [CrossRef] [PubMed]
  83. Tomasi, R.F.; Sart, S.; Champetier, T.; Baroud, C.N. Individual Control and Quantification of 3D Spheroids in a High-Density Microfluidic Droplet Array. Cell Rep. 2020, 31, 107670. [Google Scholar] [CrossRef]
  84. Ceccarelli, M.C.; Lefevre, M.C.; Marino, A.; Pignatelli, F.; Krukiewicz, K.; Battaglini, M.; Ciofani, G. Real-time monitoring of a 3D blood-brain barrier model maturation and integrity with a sensorized microfluidic device. Lab Chip 2024, 24, 5085–5100. [Google Scholar] [CrossRef]
  85. Kuhlbach, C.; da Luz, S.; Baganz, F.; Hass, V.C.; Mueller, M.M. A Microfluidic System for the Investigation of Tumor Cell Extravasation. Bioengineering 2018, 5, 40. [Google Scholar] [CrossRef]
  86. Garcia-Chame, M.; Wadhwani, P.; Pfeifer, J.; Schepers, U.; Niemeyer, C.M.; Dominguez, C.M. A Versatile Microfluidic Platform for Extravasation Studies Based on DNA Origami-Cell Interactions. Angew. Chem. Int. Ed. Engl. 2024, 63, e202318805. [Google Scholar] [CrossRef]
  87. Shen, S.; Zhang, F.; Gao, M.; Niu, Y. Concentration Gradient Constructions Using Inertial Microfluidics for Studying Tumor Cell-Drug Interactions. Micromachines 2020, 11, 493. [Google Scholar] [CrossRef]
  88. Steinberg, E.; Friedman, R.; Goldstein, Y.; Friedman, N.; Beharier, O.; Demma, J.A.; Zamir, G.; Hubert, A.; Benny, O. A fully 3D-printed versatile tumor-on-a-chip allows multi-drug screening and correlation with clinical outcomes for personalized medicine. Commun. Biol. 2023, 6, 1157. [Google Scholar] [CrossRef] [PubMed]
  89. Dadgar, N.; Gonzalez-Suarez, A.M.; Fattahi, P.; Hou, X.; Weroha, J.S.; Gaspar-Maia, A.; Stybayeva, G.; Revzin, A. A microfluidic platform for cultivating ovarian cancer spheroids and testing their responses to chemotherapies. Microsyst. Nanoeng. 2020, 6, 93. [Google Scholar] [CrossRef]
  90. Singh, D.; Deosarkar, S.P.; Cadogan, E.; Flemington, V.; Bray, A.; Zhang, J.; Reiserer, R.S.; Schaffer, D.K.; Gerken, G.B.; Britt, C.M.; et al. A microfluidic system that replicates pharmacokinetic (PK) profiles in vitro improves prediction of in vivo efficacy in preclinical models. PLoS Biol. 2022, 20, e3001624. [Google Scholar] [CrossRef] [PubMed]
  91. Lim, W.; Park, S. A Microfluidic Spheroid Culture Device with a Concentration Gradient Generator for High-Throughput Screening of Drug Efficacy. Molecules 2018, 23, 3355. [Google Scholar] [CrossRef]
  92. Lee, S.I.; Choi, Y.Y.; Kang, S.G.; Kim, T.H.; Choi, J.W.; Kim, Y.J.; Kim, T.H.; Kang, T.; Chung, B.G. 3D Multicellular Tumor Spheroids in a Microfluidic Droplet System for Investigation of Drug Resistance. Polymers 2022, 14, 3752. [Google Scholar] [CrossRef]
  93. Lipreri, M.V.; Totaro, M.T.; Boos, J.A.; Basile, M.S.; Baldini, N.; Avnet, S. A Novel Microfluidic Platform for Personalized Anticancer Drug Screening Through Image Analysis. Micromachines 2024, 15, 1521. [Google Scholar] [CrossRef] [PubMed]
  94. Maulana, T.I.; Teufel, C.; Cipriano, M.; Roosz, J.; Lazarevski, L.; van den Hil, F.E.; Scheller, L.; Orlova, V.; Koch, A.; Hudecek, M.; et al. Breast cancer-on-chip for patient-specific efficacy and safety testing of CAR-T cells. Cell Stem. Cell 2024, 31, 989–1002 e9. [Google Scholar] [CrossRef] [PubMed]
  95. Shin, S.; Choi, Y.; Jang, W.; Ulziituya, B.; Ha, G.; Kang, R.; Park, S.; Kim, M.; Zhang, Y.S.; Kim, H.J.; et al. A vascularized tumors-on-a-chip model for studying tumor-angiogenesis interplay, heterogeneity and drug responses. Mater. Today Bio 2025, 32, 101741. [Google Scholar] [CrossRef]
  96. Zhou, Y.; Wu, Y.; Paul, R.; Qin, X.; Liu, Y. Hierarchical Vessel Network-Supported Tumor Model-on-a-Chip Constructed by Induced Spontaneous Anastomosis. ACS Appl. Mater. Interfaces 2023, 15, 6431–6441. [Google Scholar] [CrossRef]
  97. Shao, S.; Delk, N.A.; Jones, C.N. A microphysiological system reveals neutrophil contact-dependent attenuation of pancreatic tumor progression by CXCR2 inhibition-based immunotherapy. Sci. Rep. 2024, 14, 14142. [Google Scholar] [CrossRef]
  98. Xu, J.; Wu, D.; Hanada, Y.; Chen, C.; Wu, S.; Cheng, Y.; Sugioka, K. Electrofluidics fabricated by space-selective metallization in glass microfluidic structures using femtosecond laser direct writing. Lab Chip 2013, 13, 4608–4616. [Google Scholar] [CrossRef] [PubMed]
  99. Misun, P.M.; Rothe, J.; Schmid, Y.R.F.; Hierlemann, A.; Frey, O. Multi-analyte biosensor interface for real-time monitoring of 3D microtissue spheroids in hanging-drop networks. Microsyst. Nanoeng. 2016, 2, 16022. [Google Scholar] [CrossRef]
  100. Neairat, T.; Al-Gawati, M.; Tul Ain, Q.; Assaifan, A.K.; Alshamsan, A.; Alarifi, A.; Alodhayb, A.N.; Alzahrani, K.E.; Albrithen, H. Development of a microcantilever-based biosensor for detecting Programmed Death Ligand 1. Saudi. Pharm. J. 2024, 32, 102051. [Google Scholar] [CrossRef]
  101. Nolan, J.K.; Nguyen, T.N.H.; Le, K.V.H.; DeLong, L.E.; Lee, H. Simple Fabrication of Flexible Biosensor Arrays Using Direct Writing for Multianalyte Measurement from Human Astrocytes. SLAS Technol. 2020, 25, 33–46. [Google Scholar] [CrossRef]
  102. Zhang, Y.S.; Aleman, J.; Shin, S.R.; Kilic, T.; Kim, D.; Mousavi Shaegh, S.A.; Massa, S.; Riahi, R.; Chae, S.; Hu, N.; et al. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc. Natl. Acad. Sci. USA 2017, 114, E2293–E2302. [Google Scholar] [CrossRef]
  103. Kanioura, A.; Filippidou, M.K.; Tsounidi, D.; Petrou, P.S.; Chatzandroulis, S.; Tserepi, A. An Organ-on-a-Chip Modular Platform with Integrated Immunobiosensors for Monitoring the Extracellular Environment. Micromachines 2025, 16, 740. [Google Scholar] [CrossRef]
  104. Morales, I.A.; Boghdady, C.M.; Campbell, B.E.; Moraes, C. Integrating mechanical sensor readouts into organ-on-a-chip platforms. Front. Bioeng. Biotechnol. 2022, 10, 1060895. [Google Scholar] [CrossRef] [PubMed]
  105. Clarke, G.A.; Hartse, B.X.; Niaraki Asli, A.E.; Taghavimehr, M.; Hashemi, N.; Abbasi Shirsavar, M.; Montazami, R.; Alimoradi, N.; Nasirian, V.; Ouedraogo, L.J.; et al. Advancement of Sensor Integrated Organ-on-Chip Devices. Sensors 2021, 21, 1367. [Google Scholar] [CrossRef] [PubMed]
  106. Fuchs, S.; Johansson, S.; Tjell, A.O.; Werr, G.; Mayr, T.; Tenje, M. In-Line Analysis of Organ-on-Chip Systems with Sensors: Integration, Fabrication, Challenges, and Potential. ACS Biomater. Sci. Eng. 2021, 7, 2926–2948. [Google Scholar] [CrossRef]
  107. Movčana, V.; Strods, A.; Narbute, K.; Rūmnieks, F.; Rimša, R.; Mozoļevskis, G.; Ivanovs, M.; Kadiķis, R.; Zviedris, K.G.; Leja, L.; et al. Organ-On-A-Chip (OOC) Image Dataset for Machine Learning and Tissue Model Evaluation. Data 2024, 9, 28. [Google Scholar] [CrossRef]
  108. Liu, B.; Qin, P.; Liu, M.; Liu, W.; Zhang, P.; Ye, Z.; Deng, Z.; Li, Z.; Gui, L. Pressure Driven Rapid Reconfigurable Liquid Metal Patterning. Micromachines 2023, 14, 717. [Google Scholar] [CrossRef]
  109. Zhang, B.; Li, J.; Zhou, J.; Chow, L.; Zhao, G.; Huang, Y.; Ma, Z.; Zhang, Q.; Yang, Y.; Yiu, C.K.; et al. A three-dimensional liquid diode for soft, integrated permeable electronics. Nature 2024, 628, 84–92. [Google Scholar] [CrossRef] [PubMed]
  110. Luo, C.; Zhang, Y.; Zhang, J.; Hui, L.; Qi, R.; Han, Y.; Sun, X.; Li, Y.; Wei, Y.; Zhang, Y.; et al. Wearable Arduino-Based Electronic Interactive Tattoo: A New Type of High-Tech Humanized Emotional Expression for Electronic Skin. Sensors 2025, 25, 2153. [Google Scholar] [CrossRef]
  111. Tao, Y.; Han, F.; Shi, C.; Yang, R.; Chen, Y.; Ren, Y. Liquid Metal-Based Flexible and Wearable Sensor for Functional Human-Machine Interface. Micromachines 2022, 13, 1429. [Google Scholar] [CrossRef]
  112. Lin, W.; Ai, L.; Wang, Y.; Yang, X.; Liao, J.; Pan, Q.; Hong, Y.; Liu, S.; Long, Z.; Khoo, B.L.; et al. Imperceptible liquid metal based tattoo for Human-Machine interface on hairy skin. Chem. Eng. J. 2024, 490, 151595. [Google Scholar] [CrossRef]
  113. Gao, Z.; Mansor, M.H.; Howard, F.; MacInnes, J.; Zhao, X.; Muthana, M. Microfluidic-Assisted Silk Nanoparticles Co-Loaded with Epirubicin and Copper Sulphide: A Synergistic Photothermal-Photodynamic Chemotherapy Against Breast Cancer. Nanomaterials 2025, 15, 221. [Google Scholar] [CrossRef]
  114. Meng, Z.; Xue, H.; Wang, T.; Chen, B.; Dong, X.; Yang, L.; Dai, J.; Lou, X.; Xia, F. Aggregation-induced emission photosensitizer-based photodynamic therapy in cancer: From chemical to clinical. J. Nanobiotechnol. 2022, 20, 344. [Google Scholar] [CrossRef] [PubMed]
  115. Zhang, T.; Deng, Y.; Liu, Y.S.; Chua, S.L.; Tang, B.Z.; Khoo, B.L. Bacterial targeted AIE photosensitizers synergistically promote chemotherapy for the treatment of inflammatory cancer. Chem. Eng. J. 2022, 447, 137579. [Google Scholar] [CrossRef]
  116. Zhang, X.Y.; Sun, K.; Abulimiti, A.; Xu, P.P.; Li, Z.Y. Microfluidic System for Observation of Bacterial Culture and Effects on Biofilm Formation at Microscale. Micromachines 2019, 10, 606. [Google Scholar] [CrossRef] [PubMed]
  117. Azharuddin, M.; Roberg, K.; Dhara, A.K.; Jain, M.V.; Darcy, P.; Hinkula, J.; Slater, N.K.H.; Patra, H.K. Dissecting multi drug resistance in head and neck cancer cells using multicellular tumor spheroids. Sci. Rep. 2019, 9, 20066. [Google Scholar] [CrossRef]
  118. van Duinen, V.; van den Heuvel, A.; Trietsch, S.J.; Lanz, H.L.; van Gils, J.M.; van Zonneveld, A.J.; Vulto, P.; Hankemeier, T. 96 perfusable blood vessels to study vascular permeability in vitro. Sci. Rep. 2017, 7, 18071. [Google Scholar] [CrossRef]
  119. Palacio-Castaneda, V.; Dumas, S.; Albrecht, P.; Wijgers, T.J.; Descroix, S.; Verdurmen, W.P.R. A Hybrid In Silico and Tumor-on-a-Chip Approach to Model Targeted Protein Behavior in 3D Microenvironments. Cancers 2021, 13, 2461. [Google Scholar] [CrossRef]
  120. Park, S.; Joo, Y.K.; Chen, Y. Versatile and High-throughput Force Measurement Platform for Dorsal Cell Mechanics. Sci. Rep. 2019, 9, 13286. [Google Scholar] [CrossRef]
  121. Bussooa, A.; Tubbs, E.; Revol-Cavalier, F.; Chmayssem, A.; Alessio, M.; Cosnier, M.-L.; Verplanck, N. Real-time monitoring of oxygen levels within thermoplastic Organ-on-Chip devices. Biosens. Bioelectron. X 2022, 11, 100198. [Google Scholar]
  122. Jang, H.; Kim, J.; Shin, J.H.; Fredberg, J.J.; Park, C.Y.; Park, Y. Traction microscopy with integrated microfluidics: Responses of the multi-cellular island to gradients of HGF. Lab Chip 2019, 19, 1579–1588. [Google Scholar] [CrossRef]
  123. Browning, A.P.; Sharp, J.A.; Murphy, R.J.; Gunasingh, G.; Lawson, B.; Burrage, K.; Haass, N.K.; Simpson, M. Quantitative analysis of tumour spheroid structure. Elife 2021, 10, e73020. [Google Scholar] [CrossRef]
  124. Soragni, C.; Vergroesen, T.; Hettema, N.; Rabussier, G.; Lanz, H.L.; Trietsch, S.J.; de Windt, L.J.; Ng, C.P. Quantify permeability using on-a-chip models in high-throughput applications. STAR Protoc 2023, 4, 102051. [Google Scholar] [CrossRef]
  125. Ahn, S.I.; Sei, Y.J.; Park, H.J.; Kim, J.; Ryu, Y.; Choi, J.J.; Sung, H.J.; MacDonald, T.J.; Levey, A.I.; Kim, Y. Microengineered human blood-brain barrier platform for understanding nanoparticle transport mechanisms. Nat. Commun. 2020, 11, 175. [Google Scholar] [CrossRef]
  126. Lin, Y.H.; Lin, C.M.; Man, K.M.; Hung, C.C.; Hsu, H.L.; Chen, Y.; Mu, H.Y.; Hsiao, T.H.; Huang, J.H. Real-time and regional analysis of the efficacy of anticancer drugs in a patient-derived intratumoral heterogeneous tumor microenvironment. Lab Chip 2025, 25, 1728–1743. [Google Scholar]
  127. Plesselova, S.; Calar, K.; Axemaker, H.; Sahly, E.; Bhagia, A.; Faragher, J.L.; Fink, D.M.; de la Puente, P. Multicompartmentalized Microvascularized Tumor-on-a-Chip to Study Tumor-Stroma Interactions and Drug Resistance in Ovarian Cancer. Cell Mol. Bioeng. 2024, 17, 345–367. [Google Scholar] [CrossRef]
  128. Ong, L.J.Y.; Chia, S.; Wong, S.Q.R.; Zhang, X.; Chua, H.; Loo, J.M.; Chua, W.Y.; Chua, C.; Tan, E.; Hentze, H.; et al. A comparative study of tumour-on-chip models with patient-derived xenografts for predicting chemotherapy efficacy in colorectal cancer patients. Front. Bioeng. Biotechnol. 2022, 10, 952726. [Google Scholar] [CrossRef]
  129. Zhou, L.; Huang, J.; Li, C.; Gu, Q.; Li, G.; Li, Z.A.; Xu, J.; Zhou, J.; Tuan, R.S. Organoids and organs-on-chips: Recent advances, applications in drug development, and regulatory challenges. Med. 2025, 6, 100667. [Google Scholar] [CrossRef]
  130. Razavi, Z.; Soltani, M.; Pazoki-Toroudi, H.; Dabagh, M. Microfluidic systems for modeling digestive cancer: A review of recent progress. Biomed. Phys. Eng. Express 2024, 10, 052002. [Google Scholar] [CrossRef]
  131. Gangwal, A.; Lavecchia, A. Artificial intelligence in preclinical research: Enhancing digital twins and organ-on-chip to reduce animal testing. Drug Discov. Today 2025, 30, 104360. [Google Scholar] [CrossRef]
  132. Moen, E.; Bannon, D.; Kudo, T.; Graf, W.; Covert, M.; Van Valen, D. Deep learning for cellular image analysis. Nat. Methods 2019, 16, 1233–1246. [Google Scholar] [CrossRef] [PubMed]
  133. Li, B.; Tang, Y.; Huang, Z.; Ma, L.; Song, J.; Xue, L. Synergistic innovation in organ-on-a-chip and organoid technologies: Reshaping the future of disease modeling, drug development and precision medicine. Protein. Cell 2025. [Google Scholar] [CrossRef]
  134. Stringer, C.; Wang, T.; Michaelos, M.; Pachitariu, M. Cellpose: A generalist algorithm for cellular segmentation. Nat. Methods 2021, 18, 100–106. [Google Scholar]
  135. Holzreuter, M.A.; Segerink, L.I. Innovative electrode and chip designs for transendothelial electrical resistance measurements in organs-on-chips. Lab Chip 2024, 24, 1121–1134. [Google Scholar] [CrossRef]
  136. Deng, S.; Li, C.; Cao, J.; Cui, Z.; Du, J.; Fu, Z.; Yang, H.; Chen, P. Organ-on-a-chip meets artificial intelligence in drug evaluation. Theranostics 2023, 13, 4526. [Google Scholar] [CrossRef]
  137. Li, J.; Chen, J.; Bai, H.; Wang, H.; Hao, S.; Ding, Y.; Peng, B.; Zhang, J.; Li, L.; Huang, W. An overview of organs-on-chips based on deep learning. Research 2022, 2022, 9869518. [Google Scholar] [CrossRef]
  138. Ronneberger, O.; Fischer, P.; Brox, T. In U-net: Convolutional networks for biomedical image segmentation. In Proceedings of the 18th International Conference on Medical Image Computing and Computer-Assisted Intervention, Munich, Germany, 5–9 October 2015; Springer: Cham, Switzerland, 2015; pp. 234–241. [Google Scholar]
  139. Caicedo, J.C.; Cooper, S.; Heigwer, F.; Warchal, S.; Qiu, P.; Molnar, C.; Vasilevich, A.S.; Barry, J.D.; Bansal, H.S.; Kraus, O.; et al. Data-analysis strategies for image-based cell profiling. Nat. Methods 2017, 14, 849–863. [Google Scholar]
  140. Sabaté Del Río, J.; Ro, J.; Yoon, H.; Park, T.E.; Cho, Y.K. Integrated technologies for continuous monitoring of organs-on-chips: Current challenges and potential solutions. Biosens. Bioelectron. 2023, 224, 115057. [Google Scholar] [CrossRef]
  141. Chen, F.; Zhang, H.Y.; Wan, Y.L.; Jia, J.N.; Wang, R.Z.; Gao, C.; Chao, Z.Y.; Ru, Y.H.; Wang, Z.; Cheng, K.; et al. Artificial intelligence-assisted organoid construction in congenital heart disease: Current applications and future prospects. Front. Bioeng. Biotechnol. 2025, 13, 1691972. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Design and fabrication of the mechano-OoC platforms. (A) Colorectal tumor-on-chip design and configuration comprises a SolidWorks-designed (SolidWorks 2024) 3D-printed mini-reactor with defined shape and dimensions, Caco2 microtumors in hydrogel microspheres fabricated by dripping bioink (3.5% alginate, 2.5% GelMA, 1.5 × 106 cells/mL) into 4% CaCl2 and 12 g/L Tween 80 bath, and a closed system via acrylic lid sealing and connection to syringe pump and waste tube [48]. (B) The design and fabrication of the organoid-on-chip device involve AutoCAD-designed top (AutoCAD v25.1) (one inlet channel splitting into four sub-channels and one outlet) and bottom (four 6-mm-diameter round wells for organoid seeding/growth) layers, PMMA molds fabricated via milling machine for producing corresponding PDMS layers, and subsequent device setup [51]. (C) The figure includes an image of the LumenX DLP printer (left), a schematic of a basic DLP printer in operation (middle), and a CAD model for printing vertically oriented channels of varying sizes (right) [53]. (D) Printable PEGDA channel diameter varies with single-component adjustments (LAP/tartrazine concentration, projector power); microscope images of 0.1–1.0 mm channels and fluorescent dye confirm hollowness [53].
Figure 1. Design and fabrication of the mechano-OoC platforms. (A) Colorectal tumor-on-chip design and configuration comprises a SolidWorks-designed (SolidWorks 2024) 3D-printed mini-reactor with defined shape and dimensions, Caco2 microtumors in hydrogel microspheres fabricated by dripping bioink (3.5% alginate, 2.5% GelMA, 1.5 × 106 cells/mL) into 4% CaCl2 and 12 g/L Tween 80 bath, and a closed system via acrylic lid sealing and connection to syringe pump and waste tube [48]. (B) The design and fabrication of the organoid-on-chip device involve AutoCAD-designed top (AutoCAD v25.1) (one inlet channel splitting into four sub-channels and one outlet) and bottom (four 6-mm-diameter round wells for organoid seeding/growth) layers, PMMA molds fabricated via milling machine for producing corresponding PDMS layers, and subsequent device setup [51]. (C) The figure includes an image of the LumenX DLP printer (left), a schematic of a basic DLP printer in operation (middle), and a CAD model for printing vertically oriented channels of varying sizes (right) [53]. (D) Printable PEGDA channel diameter varies with single-component adjustments (LAP/tartrazine concentration, projector power); microscope images of 0.1–1.0 mm channels and fluorescent dye confirm hollowness [53].
Ijms 27 01330 g001
Figure 2. The design of microfluidic perfusion chips and their application in drug screening. (A) Microfluidic device mixing effect (20 μL min−1, successive spiral mixer regulation) involves device simulation imaging, with three areas (start, center, end) in three mixers for mixing status identification [87]. (B) Development and characterization of a spheroid-containing microfluidic platform involve PDMS-glass-agarose (cone-shaped wells) structure, assembled device images, and dye/OrganoFlow® validation of no leakage and normal fluid flow [93].
Figure 2. The design of microfluidic perfusion chips and their application in drug screening. (A) Microfluidic device mixing effect (20 μL min−1, successive spiral mixer regulation) involves device simulation imaging, with three areas (start, center, end) in three mixers for mixing status identification [87]. (B) Development and characterization of a spheroid-containing microfluidic platform involve PDMS-glass-agarose (cone-shaped wells) structure, assembled device images, and dye/OrganoFlow® validation of no leakage and normal fluid flow [93].
Ijms 27 01330 g002
Figure 3. Fabrication processes and dimensional details relevant to the pressure-driven rapid reconfigurable liquid metal patterning strategy. (A) Standard soft lithography for PDMS component fabrication, (B,C) preparation of single- and double-pattern chips, (D) preparation of dual-frequency reconfigurable antennas, (E) and specific dimensional parameters of liquid metal patterns formed by elastic polymer film deformation in the “pattern—film—cavity” sandwich structure under working medium pressure [108].
Figure 3. Fabrication processes and dimensional details relevant to the pressure-driven rapid reconfigurable liquid metal patterning strategy. (A) Standard soft lithography for PDMS component fabrication, (B,C) preparation of single- and double-pattern chips, (D) preparation of dual-frequency reconfigurable antennas, (E) and specific dimensional parameters of liquid metal patterns formed by elastic polymer film deformation in the “pattern—film—cavity” sandwich structure under working medium pressure [108].
Ijms 27 01330 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, L.; Cheung, J.C.W.; Zhao, X.; Khoo, B.L.; Wong, S.H.D. Mechano-Organ-on-Chip for Cancer Research. Int. J. Mol. Sci. 2026, 27, 1330. https://doi.org/10.3390/ijms27031330

AMA Style

Wang L, Cheung JCW, Zhao X, Khoo BL, Wong SHD. Mechano-Organ-on-Chip for Cancer Research. International Journal of Molecular Sciences. 2026; 27(3):1330. https://doi.org/10.3390/ijms27031330

Chicago/Turabian Style

Wang, Luyang, James Chung Wai Cheung, Xia Zhao, Bee Luan Khoo, and Siu Hong Dexter Wong. 2026. "Mechano-Organ-on-Chip for Cancer Research" International Journal of Molecular Sciences 27, no. 3: 1330. https://doi.org/10.3390/ijms27031330

APA Style

Wang, L., Cheung, J. C. W., Zhao, X., Khoo, B. L., & Wong, S. H. D. (2026). Mechano-Organ-on-Chip for Cancer Research. International Journal of Molecular Sciences, 27(3), 1330. https://doi.org/10.3390/ijms27031330

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop