Cancer Letters

Cancer Letters

Volume 414, 1 February 2018, Pages 174-180
Cancer Letters

Mini-review
Organoids: An intermediate modeling platform in precision oncology

https://doi.org/10.1016/j.canlet.2017.11.021Get rights and content

Highlights

  • Organoids serve as an intermediate modeling platform in precision oncology.

  • Organoids have potential translational applications for disease modeling, drug screening, and chemosensitivity studies.

  • The marriage of gene editing system and organoids can enhance cancer modeling and clinical applications.

Abstract

Cancer harbors variable heterogeneity and plasticity. Thus far, our comprehension is greatly based on cell lines, organoids, and patient-derived tumor xenografts (PDTXs). Organoids are a three-dimensional in vitro culture platform constructed from self-organizing stem cells. They can almost accurately recapitulate tumor heterogeneity and microenvironment “in a dish,” which surpass established cell lines and are not as expensive and time-consuming as PDTXs. As an intermediate model, tumor organoids are also used to study the fundamental issues of tumorigenesis and metastasis. They are specifically applied for drug testing and stored as “living biobanks.” In this review, we highlight the translational applications of organoid technologies in tumor research and precision medicine, discuss the advantages and limitations compared with other mentioned methods, and provide our outlook on its future.

Introduction

Cancer causes a significant number of deaths worldwide. Although the last two decades have witnessed a 25% decline in cancer death rates that is equivalent to 2.1 million survivors [1], the absolute number of US annual cancer incidence and related deaths is projected to increase by 2030 [2]. Surgery, chemotherapy, and radiotherapy are routinely used to treat cancer together with immunotherapy and targeted therapy. However, they are not effective enough. Inter-patient heterogeneity varies the curative effects from person to person [3]. Researches use cell lines and patient-derived tumor xenografts (PDTXs) to study tumorigenesis. However, these methods have limitations. Genetic drifts and two-dimensional (2D) vision make the findings of cell lines unrealistic and unreliable; meanwhile, using PDTXs is expensive and time-consuming (Fig. 1a) [4]. These obstacles thus help drive the development of tumor organoids, which can recapitulate tumor biology relatively accurately.

Not until the recent decade did this three-dimensional (3D) in vitro culture technology stage a comeback (Fig. 2). Nowadays, organoid techniques provide unique platforms to model organ or tissue development and human diseases. Organoids are derived from pluripotent stem cells (PSCs) and organ-restricted adult stem cells (ASCs); the former is composed of pluripotent embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) [5]. Organoids can also be generated from established cell lines and transformed into PDTXs (Fig. 1b) [4].

Derived from patients, organoids are crucial tools for disease modeling, including cancer, cystic fibrosis (CF), and Zika virus [6]; neurodevelopmental disorders [7], [8], [9], [10]; liver disorders [11]; and drug testing [12], [13]; moreover, they can be stored as “living biobanks” (Fig. 1c) [14], [15]. Recently, the success of applying the prokaryotic clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated protein 9 (Cas9) system to correct the CFTR locus made treating future CF patients possible [16]; meanwhile, established tumor-derived organoid cultures can be used for high-throughput drug screening [14]. These findings pave the way for the development of precision medicine. In this review, we mainly focus on organoids as a technology platform for cancer precision medicine, especially with potential translational applications for disease modeling, drug screening, and chemosensitivity studies.

Section snippets

Resources and methods

Methods for developing 3D organoids were based from earlier 2D culture systems and have improvements. Briefly, tumor cells isolated from tumor samples or PSCs/ASCs are embedded in serum-free media with growth factors to form organoids [17], [18]. The introduction of components, including growth factor-optimized media and basement membrane matrix (Matrigel), gives organoids the ability to self-renew and differentiate. Though different laboratories have their own preferences, Matrigel and

Advantages and limitations vs cell lines and PDTXs

As mentioned before, unlike traditional in vitro 2D cultures, organoids are similar to original tissues in their genetic composition and cellular architecture, harboring small populations of genetically stable, self-renewing stem-like cells that can give rise to fully differentiated progeny, comprising major cell lineages at frequencies similar to those in living tissue [73].

Cell lines have been a widely-used model for cellular studies for a long time. They are easy to culture and beneficial

Outlook and conclusions

Three-dimensional organoids are becoming popular and significant in tumor research and precision medicine. Three-dimensional bioprinting of multiple 3D organoids can be connected with a vascular network to develop an “organ-on-a-chip”, which may lead to advancements in toxicology. It enables us to observe pharmacokinetics and pharmacodynamics [70].

Transplantation of engineered patient-derived organoids will be potentially applied in regenerative medicine, but how to prepare their

Conflicts of interest

The authors declare that they have no competing interests.

Acknowledgements

We apologize to those colleagues whose important work could not be cited due to space constraints. This work was in part supported by the National Key Research and Development Program of China (No. 2017FYA0205300), the National Natural Science Foundation of China (No. 81702944, 81272801), Special Presidential Foundation of General Hospital of Jinan Military Command (No.2016BS04) and Shanghai Jiao Tong University Med-X Fund (No.YG2015MS20).

References (86)

  • L.A. Baker et al.

    Modeling pancreatic cancer with organoids

    Trends Cancer

    (2016)
  • D. Loessner et al.

    Engineered microenvironments provide new insights into ovarian and prostate cancer progression and drug responses

    Adv. Drug Deliv. Rev.

    (2014)
  • B. Ponsioen et al.

    Cancer Systems Biology: live imaging of intestinal tissue in health and disease

    Curr. Opin. Syst. Biol.

    (2017)
  • H. Endo et al.

    Spheroid culture of primary lung cancer cells with neuregulin 1/HER3 pathway activation

    J. Thorac. Oncol.

    (2013)
  • P. Codega et al.

    Dissecting glioma invasiveness in a 3D-organotypic model

    Trends Mol. Med.

    (2017)
  • J.K.C. Chuah et al.

    Stem cell-derived kidney cells and organoids: recent breakthroughs and emerging applications

    Biotechnol. Adv.

    (2017)
  • M.E. Todhunter et al.

    Cell and tissue biology paves a path to breast cancer prevention

    Trends Cancer

    (2017)
  • D. Hanahan et al.

    Hallmarks of cancer: the next generation

    Cell

    (2011)
  • M. Fujii et al.

    A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis

    Cell Stem Cell

    (2016)
  • D. Gao et al.

    Organoid cultures derived from patients with advanced prostate cancer

    Cell

    (2014)
  • A. Kaisani et al.

    Branching morphogenesis of immortalized human bronchial epithelial cells in three-dimensional culture

    Differentiation

    (2014)
  • R.R. Nadkarni et al.

    Organoids as a model system for studying human lung development and disease

    Biochem. Biophys. Res. Commun.

    (2016)
  • R.J. Platt et al.

    CRISPR-Cas9 knockin mice for genome editing and cancer modeling

    Cell

    (2014)
  • C. Unger et al.

    Modeling human carcinomas: physiologically relevant 3D models to improve anti-cancer drug development

    Adv. drug Deliv. Rev.

    (2014)
  • Z. Liu et al.

    Lessons learned from two decades of anticancer drugs

    Trends Pharmacol. Sci.

    (2017)
  • W. Peng et al.

    3D bioprinting for drug discovery and development in pharmaceutics

    Acta Biomater.

    (2017)
  • D. Gao et al.

    Organoid development in cancer genome discovery

    Curr. Opin. Genet. Dev.

    (2015)
  • N. Sachs et al.

    Organoid cultures for the analysis of cancer phenotypes

    Curr. Opin. Genet. Dev.

    (2014)
  • P.J. Schweiger et al.

    Modeling human disease using organotypic cultures

    Curr. Opin. Cell Biol.

    (2016)
  • D. Behrens et al.

    Pancreatic cancer models for translational research

    Pharmacol. Ther.

    (2017)
  • K. Schenke-Layland et al.

    In vitro human tissue models—moving towards personalized regenerative medicine

    Adv. drug Deliv. Rev.

    (2011)
  • M.T. Makale et al.

    The autonomic nervous system and cancer

    Biocybern. Biomed. Eng.

    (2017)
  • H. Venkatesh et al.

    Neuronal activity in ontogeny and oncology

    Trends Cancer

    (2017)
  • A.C. Wan

    Recapitulating cell–cell interactions for organoid construction–are biomaterials dispensable?

    Trends Biotechnol.

    (2016)
  • R.L. Siegel et al.

    Cancer statistics

    CA Cancer J. Clin.

    (2017)
  • L. Rahib et al.

    Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States

    Cancer Res.

    (2014)
  • S. Wang et al.

    The potential of organoids in urological cancer research. Nature reviews

    Urology

    (2017)
  • J.A. Bagley et al.

    Fused cerebral organoids model interactions between brain regions

    Nat. Meth.

    (2017)
  • G. Quadrato et al.

    The promises and challenges of human brain organoids as models of neuropsychiatric disease

    Nat. Med.

    (2016)
  • H.E. Francies et al.

    Drug sensitivity assays of human cancer organoid cultures

  • M.A. Cantrell et al.

    Organoid modeling for cancer precision medicine

    Genome Med.

    (2015)
  • F.S. Collins et al.

    A new initiative on precision medicine

    N. Engl. J. Med.

    (2015)
  • C. Pauli et al.

    Personalized in vitro and in vivo cancer models to guide precision medicine

    Cancer Discov.

    (2017)
  • Cited by (57)

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text