Elsevier

Biomaterials

Volume 149, December 2017, Pages 98-115
Biomaterials

Review
Organ-on-chip models of cancer metastasis for future personalized medicine: From chip to the patient

https://doi.org/10.1016/j.biomaterials.2017.10.005Get rights and content

Abstract

Most cancer patients do not die from the primary tumor but from its metastasis. Current in vitro and in vivo cancer models are incapable of satisfactorily predicting the outcome of various clinical treatments on patients. This is seen as a serious limitation and efforts are underway to develop a new generation of highly predictive cancer models with advanced capabilities. In this regard, organ-on-chip models of cancer metastasis emerge as powerful predictors of disease progression. They offer physiological-like conditions where the (hypothesized) mechanistic determinants of the disease can be assessed with ease. Combined with high-throughput characteristics, the employment of organ-on-chip technology would allow pharmaceutical companies and clinicians to test new therapeutic compounds and therapies. This will permit the screening of a large battery of new drugs in a fast and economic manner, to accelerate the diagnosis of the disease in the near future, and to test personalized treatments using cells from patients. In this review, we describe the latest advances in the field of organ-on-chip models of cancer metastasis and their integration with advanced imaging, screening and biosensing technologies for future precision medicine applications. We focus on their clinical applicability and market opportunities to drive us forward to the next generation of tumor models for improved cancer patient theranostics.

Introduction

Most cancer deaths result from metastasis. Metastasis is defined as the sequence of events leading to the spread of cancer cells from the site of primary tumor to new locations in the human body. A lot of investigation is being carried out into the molecular basis of tumor growth and dissemination [1]. However, the mechanism of metastasis is still poorly understood [2], [3]. In addition to genetic and external environmental factors, tumor expansion is also determined by the structural properties of the tumor microenvironment [4]. The details on the mechanism of conversion of a physical stimulus in the tumor microenvironment, such as cell-cell/extracellular matrix (ECM) interactions or fluid shear forces, into a biochemical response during tumor progression are not well understood. Thus, the comprehension of the disease and its progression into metastasis is still limited [5], [6], [7], [8]. Metastasis is also related to mechanism of drug resistance which still remains unclear [9], [10], [11]. Multiple efforts are directed toward the development of cancer metastasis models that can help in understanding the disease and in the development of innovative therapeutic strategies. Traditionally, standard in vivo and in vitro models are used to elucidate the mechanisms involved in metastasis [12], [13], [14], [15], [16], [17]. The complexity encountered in humans is reproduced with higher fidelity using in vivo models. However, the individual contribution of interconnected physical and biochemical parameters in an in vivo model cannot be assessed easily. In addition, they fail in predicting the clinical efficacy of new drug candidates [18], [19]. The in vitro, though fully controllable, lack the hierarchical complexity of the tumor niche needed to reproduce the native scenario. This threatens the relevance of the data obtained using in vitro models. Pre-clinical experimentation demands highly sophisticated and physiologically relevant in vitro models capable of recapitulating both the biomolecular and structural properties of the tumor niche along with the dynamic events occurring during the propagation of cancer. In this regard, organ-on-chip technology can be used to design biomimetic microfluidic devices containing human cells to replicate fundamental functional units of human tissues and organs in vitro [20], [21], [22], [23]. Applied to cancer research, organ-on-chip models of cancer metastasis, or metastasis-on-chip (MoC) emerge as a promising methodology for studying the disease under physiological-like conditions [24].

MoC models provide multiple advantages compared to their in vitro and in vivo counterparts, which have many barriers for their clinical applications. During the last decade, MoC models have been used to study the contribution of the mechanical and biochemical cues in tumor dissemination, including the impact of cell-cell/stroma interactions [12], [25], [26], [27], or cytokine gradients [28], among others. Similarly, several events described in the metastasis cascade have been successfully reproduced. This includes cancer cell invasion, angiogenesis, intravasation, extravasation, colonization, and most importantly, the impact of circulating tumor cells (CTCs) in tumor dissemination. Several MoC models have been developed toward their early detection, capturing, analysis and use for exploring their diagnostic and prognostic potential [29]. This is critically important because CTCs are responsible for most cancer-related deaths [30], [31], [32], [33].

This review describes the latest and most relevant advances in MoC models, their main advantages, limitations, and future perspectives for cancer research. We anticipate that this new generation of tumor models will provide new insights into the molecular and mechanical mechanisms of metastasis, which have remained elusive due to the limitations of current models. Further, when combined with advanced imaging, sensing and screening techniques, MoC models may be employed to understand the mechanism of targeted drug delivery in vitro, improving the knowledge on current treatments and potentially developing new therapeutic avenues [34]. Finally, the translation of MoC models into the pharmaceutical and healthcare market will require MoC models to display unprecedented capabilities. This new generation of tumor models will univocally contribute to improve the prognosis of cancer patients reducing the cost in the healthcare system.

Section snippets

Current cancer metastasis models: an overview

A large plethora of cancer metastasis models have been described in the literature during the last years. These standard models include both experimental (in vitro and in vivo) and computational (in silico) approaches, which have provided valuable insights into the mechanistic determinants of the disease. They have been extremely useful for studying tumor dissemination, screening anti-cancerous drugs, or testing novel therapeutic strategies. However, they also display serious limitations. In

Microfluidic models of cancer metastasis: metastasis-on-chip

Cancer metastasis involves a complex cascade of events (Fig. 2a) [3]. Typically, this cascade is first initiated by an uncontrolled growth of the primary malignant tumor followed by the invasion of cancer cells into the surrounding stroma. Next, a deficient oxygen supply serves as a cue for the secretion of cytokines and growth factors which induce the formation of new microvasculature – angiogenesis – that aids in the transmigration of the cancer cells, as well as ctDNA/RNA, exosomes, or

Integration of advanced imaging, screening, and biosensing technologies into organ-on-chip devices: towards precision medicine

The real-time monitoring of tumor growth and progression is challenging and difficult to accomplish with standard methodologies. Genotyping tumor tissue in search of genetic alterations is a routine practice in clinical oncology to provide physicians with highly valuable information. However, the extracted biopsy only represents a single snapshot in time and is subjected to selection bias also due to tumor heterogeneity. Multiple or serial biopsies in time may provide valuable information about

Personalized patient-derived MoC models for clinical applications

Future point-of-care MoC models must fulfill rigid requirements if intended for clinical applications. First, they need to reproduce the complexity of each individual patient to stand for the genetic heterogeneity of tumors and at the same time maintaining this complexity low to keep the model clinically-relevant. This implies the use of engineered biomaterials and cells from patients to develop personalized models of cancer metastasis [142], [143]. Second, they must integrate multi-organ

Conclusions

Standard in vitro and in vivo models of cancer metastasis display certain limitations which limit their applicability in the clinics. The recent progress in bioengineering techniques and microfabrication tools allows the production of a new generation of cancer metastasis models based on organ-on-chip technology. Different events in the metastasis cascade have been successfully reproduced, including organ-specificity and hybrid models. These metastasis-on-chip devices can provide the needed

Acknowledgements

The authors acknowledge the financial support from the European Union Framework Programme for Research and Innovation Horizon 2020 on Forefront Research in 3D Disease Cancer Models as in vitro Screening Technologies (FoReCaST) under grant agreement no 668983. Conflicts of interest: none.

References (148)

  • L.-B. Weiswald et al.

    Spherical cancer models in tumor biology

    Neoplasia

    (2015)
  • S. Levinson et al.

    Drosophila cancer models identify functional differences between ret fusions

    Cell Rep.

    (2016)
  • J. Yen et al.

    Zebrafish models of cancer: progress and future challenges

    Curr. Opin. Genet. Dev.

    (2014)
  • M. Sonoshita et al.

    Chapter nine - modeling human cancers in Drosophila

  • A. Banerjee et al.

    Mutations and interactions in human ERalpha and bZIP proteins: an in silico approach for cell signaling in breast oncology

    Gene

    (2017)
  • A.T. Stratmann et al.

    Establishment of a human 3D lung cancer model based on a biological tissue matrix combined with a Boolean in silico model

    Mol. Oncol.

    (2014)
  • M.A. Al-Mamun et al.

    An in silico model to demonstrate the effects of Maspin on cancer cell dynamics

    J. Theor. Biol.

    (2016)
  • C. Eurtivong et al.

    3-Amino-thieno[2,3-b]pyridines as microtubule-destabilising agents: molecular modelling and biological evaluation in the sea urchin embryo and human cancer cells

    Bioorg. Med. Chem.

    (2017)
  • H. Somaweera et al.

    A review of chemical gradient systems for cell analysis

    Anal. Chim. Acta

    (2016)
  • L.L. Bischel et al.

    Tubeless microfluidic angiogenesis assay with three-dimensional endothelial-lined microvessels

    Biomaterials

    (2013)
  • B. Kwak et al.

    Simulation of complex transport of nanoparticles around a tumor using tumor-microenvironment-on-chip

    J. Control Release

    (2014)
  • S. Kim et al.

    Three-dimensional biomimetic model to reconstitute sprouting lymphangiogenesis in vitro

    Biomaterials

    (2016)
  • J. Massague et al.

    Metastatic colonization by circulating tumour cells

    Nature

    (2016)
  • A.S. Azevedo et al.

    Metastasis of circulating tumor cells: favorable soil or suitable biomechanics, or both?

    Cell Adh Mig

    (2015)
  • K.M. Stroka et al.

    Physical Biology in Cancer. 4. Physical cues guide tumor cell adhesion and migration

    Am. J. Physiol. Cell Physiol.

    (2014)
  • J.S. Zawistowski et al.

    Enhancer remodeling during adaptive bypass to MEK inhibition is attenuated by pharmacologic targeting of the P-TEFb complex

    Cancer Discov.

    (2017)
  • D. Brunen et al.

    Drug therapy: exploiting synthetic lethality to improve cancer therapy

    Nat. Rev. Clin. Oncol.

    (2017)
  • V. Lopes-Rodrigues et al.

    Identification of the metabolic alterations associated with the multidrug resistant phenotype in cancer and their intercellular transfer mediated by extracellular vesicles

    Sci. Rep.

    (2017)
  • M.E. Katt et al.

    In vitro tumor models: advantages, disadvantages, variables, and selecting the right platform

    Front. Bioeng. Biotechnol.

    (2016)
  • P. Benien et al.

    3D tumor models: history, advances and future perspectives

    Future Oncol.

    (2014)
  • M. Zanoni et al.

    3D tumor spheroid models for in vitro therapeutic screening: a systematic approach to enhance the biological relevance of data obtained

    Sci. Rep.

    (2016)
  • S. Yang et al.

    Mouse models for tumor metastasis

  • W.O. Miles et al.

    Modeling tumor invasion and metastasis in Drosophila

    Dis. Model Mech.

    (2011)
  • G. Caponigro et al.

    Advances in the preclinical testing of cancer therapeutic hypotheses

    Nat. Rev. Drug Discov.

    (2011)
  • M. Hay et al.

    Clinical development success rates for investigational drugs

    Nat. Biotech.

    (2014)
  • D. Huh et al.

    Microfabrication of human organs-on-chips

    Nat. Protoc.

    (2013)
  • A. Skardal et al.

    A reductionist metastasis-on-a-chip platform for in vitro tumor progression modeling and drug screening

    Biotech. Bioeng.

    (2016)
  • I. Maschmeyer et al.

    A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents

    Lab. Chip

    (2015)
  • J.D. Caplin et al.

    Microfluidic organ-on-a-chip technology for advancement of drug development and toxicology

    Adv. Healthc. Mater

    (2015)
  • E.W. Esch et al.

    Organs-on-chips at the frontiers of drug discovery

    Nat. Rev. Drug Discov.

    (2015)
  • S.N. Bhatia et al.

    Microfluidic organs-on-chips

    Nat. Biotech.

    (2014)
  • R. Portillo-Lara et al.

    Microengineered cancer-on-a-chip platforms to study the metastatic microenvironment

    Lab. Chip

    (2016)
  • R. Riahi et al.

    A microfluidic model for organ-specific extravasation of circulating tumor cells

    Biomicrofluidics

    (2014)
  • J.W. Neal et al.

    Developing biomarker-specific end points in lung cancer clinical trials

    Nat. Rev. Clin. Oncol.

    (2015)
  • A. Toss et al.

    CTC enumeration and characterization: moving toward personalized medicine

    Ann. Transl. Med.

    (2014)
  • C. Wittekind et al.

    Cancer invasion and metastasis

    Oncology

    (2005)
  • S.H. Au et al.

    Clusters of circulating tumor cells traverse capillary-sized vessels

    Proc. Natl. Acad. Sci. U. S. A.

    (2016)
  • K. Kingwell

    3D cell technologies head to the R&D assembly line

    Nat. Rev. Drug Discov.

    (2017)
  • D.W. Hutmacher

    Biomaterials offer cancer research the third dimension

    Nat. Mater

    (2010)
  • S. Zhou et al.

    Substrates with patterned topography reveal metastasis of human cancer cells

    Biomed. Mater

    (2017)
  • Cited by (143)

    View all citing articles on Scopus
    View full text