AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (581.9 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Organoid models of gastrointestinal Neoplasms: Origin, current status and future applications in personalized medicine

Yi Pan,Shuliang Zhao( )Zhijun Cao( )
Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, 145 Middle Shandong Road, Shanghai, 200001, China

Peer review under responsibility of Chongqing Medical University.

Show Author Information

Abstract

The in vitro organoid model is a major technological breakthrough that has been established as an important tool in many basic biological and clinical applications. This near-physiological 3D culture system accurately models various biological processes, including tissue renewal, stem cell/niche functions and tissue responses to drugs, mutations or damage. Organoids have the potential value of being an accurate model for disease predictions or drug screening applications and to identify the ideal treatment for that patient. Carcinogenesis can be modeled by mutating specific cancer genes in wild-type organoids; and patient-derived organoids provide an important resource in the development of personalized cancer treatment. Organoids from cancer patients could be used to identify the ideal treatment for a specific patient by growing matched healthy and diseased organoids from human cancer patients which additionally enables clinical screens for drug combinations. Organoids could also provide autologous cells or—in the future—tissue for transplantation. In this review, we discuss the current advances, challenges and potential applications of this technique in gastrointestinal neoplasms.

References

1

Hayflick L. The limited in vitro lifetime of human diploid cell strains. Exp Cell Res. 1965;37:614-636.

2

Huch M, Gehart H, van Boxtel R, et al. Long-term culture of genome-stable bipotent stem cells from adult human liver. Cell. 2015;160:299-312.

3

Smith E, Cochrane WJ. Cystic organoid teratoma: (report of a case). Can Med Assoc J. 1946;55:151-152.

4

Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345:124-125.

5

Weiss P, Taylor AC. Reconstitution of complete organs from single-cell suspensions of chick embryos in advanced stages of differentiation. Proc Natl Acad Sci USA. 1960;46:1177-1185.

6

Barker N, van Es JH, Kuipers J, et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature. 2007;449:1003-1007.

7

Korinek V, Barker N, Moerer P, et al. Depletion of epithelial stem-cell compartments in the small intestine of mice lacking Tcf-4. Nat Genet. 1998;19:379-383.

8

Pinto D, Gregorieff A, Begthel H, Clevers H. Canonical Wnt signals are essential for homeostasis of the intestinal epithelium. Genes Dev. 2003;17:1709-1713.

9

de Lau W, Barker N, Low TY, et al. Lgr5 homologues associate with Wnt receptors and mediate R-spondin signalling. Nature. 2011;476:293-297.

10

Kim KA, Kakitani M, Zhao J, et al. Mitogenic influence of human R-spondin1 on the intestinal epithelium. Science. 2005;309:1256-1259.

11

Clevers H. The intestinal crypt, a prototype stem cell compartment. Cell. 2013;154:274-284.

12

Haramis AP, Begthel H, van den Born M, et al. De novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine. Science. 2004;303:1684-1686.

13

Wong VW, Stange DE, Page ME, et al. Lrig1 controls intestinal stem-cell homeostasis by negative regulation of ErbB signalling. Nat Cell Biol. 2012;14:401-408.

14

Hofmann C, Obermeier F, Artinger M, et al. Cell-cell contacts prevent anoikis in primary human colonic epithelial cells. Gastroenterology. 2007;132:587-600.

15

Jensen KB, Driskell RR, Watt FM. Assaying proliferation and differentiation capacity of stem cells using disaggregated adult mouse epidermis. Nat Protoc. 2010;5:898-911.

16

Montesano R, Schaller G, Orci L. Induction of epithelial tubular morphogenesis in vitro by fibroblast-derived soluble factors. Cell. 1991;66:697-711.

17

Xu C, Inokuma MS, Denham J, et al. Feeder-free growth of undifferentiated human embryonic stem cells. Nat Biotechnol. 2001;19:971-974.

18

Grun D, Lyubimova A, Kester L, et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature. 2015;525:251-255.

19

Sato T, Vries RG, Snippert HJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262-265.

20

Sato T, Stange DE, Ferrante M, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and barrett's epithelium. Gastroenterology. 2011;141:1762-1772.

21

Jung P, Sato T, Merlos-Suarez A, et al. Batlle EIsolation and in vitro expansion of human colonic stem cells. Nat Med. 2011;17:1225-1227.

22

Fordham RP, Yui S, Hannan NR, et al. Transplantation of expanded fetal intestinal progenitors contributes to colon regeneration after injury. Cell Stem Cell. 2013;13:734-744.

23

Lugli N, Kamileri I, Keogh A, et al. R-spondin 1 and noggin facilitate expansion of resident stem cells from non-damaged gallbladders. EMBO Rep. 2016;17:769-779.

24

Huch M, Dorrell C, Boj SF, et al. In vitro expansion of single Lgr5+ liver stem cells induced by Wnt-driven regeneration. Nature. 2013;494:247-250.

25

Boj SF, Hwang CI, Baker LA, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324-338.

26

Huch M, Bonfanti P, Boj SF, et al. Unlimited in vitro expansion of adult bi-potent pancreas progenitors through the Lgr5/R-spondin axis. EMBO J. 2013;32:2708-2721.

27

BarkerN, Huch M, Kujala P, et al. Lgr5(+ve) stem cells drive self-renewal in the stomach and build long-lived gastric units in vitro. Cell Stem Cell. 2010;6:25-36.

28

Bartfeld S, Bayram T, van de Wetering M, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148:126-136. e126.

29

Stange DE, Koo BK, Huch M, et al. Differentiated Troy+ chief cells act as reserve stem cells to generate all lineages of the stomach epithelium. Cell. 2013;155:357-368.

30

Sato T, van Es JH, Snippert HJ, et al. Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts. Nature. 2011;469:415-418.

31

Bi T, Lan H, Hu X, et al. Antitumor effect of FP3 on a patient-derived tumor tissue xenograft model of rectal carcinoma. Hepatogastroenterology. 2013;60:1950-1954.

32

Smith NR, Baker D, Farren M, et al. Tumor stromal architecture can define the intrinsic tumor response to VEGF-targeted therapy. Clin Cancer Res. 2013;19:6943-6956.

33

Schumacher MA, Aihara E, Feng R, et al. The use of murine-derived fundic organoids in studies of gastric physiology: the use of fundic organoids. J Physiol. 2015;593:1809-1827 [a].

34

Bertaux-Skeirik N, Feng R, Schumacher MA, et al. CD44 plays a functional role in Helicobacter pylori-induced epithelial cell proliferation. PLoS Pathog. 2015;11:e1004663.

35

Schlaermann P, Toelle B, Berger H, et al. A novel human gastric primary cell culture system for modelling Helicobacter pylori infection in vitro. Gut. 2016;65:202-213.

36

Van de Wetering M, Francies HE, Francis JM, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161:933-945.

37

Pompaiah M, Bartfeld S. Gastric organoids: an emerging model system to study helicobacter pylori pathogenesis. Curr Top Microbiol Immunol. 2017;400:149-168.

38

Huang L, Holtzinger A, Jagan I, et al. Ductal pancreatic cancer modeling and drug screening using human pluripotent stem cell- and patient-derived tumor organoids. Nat Med. 2015;21:1364-1371.

39

Kim J, Hoffman JP, Alpaugh RK, et al. An IPSC line from human pancreatic ductal adenocarcinoma undergoes early to invasive stages of pancreatic cancer progression. Cell Rep. 2013;3:2088-2099.

40

Kim J, Bamlet WR, Oberg AL, et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and ca19-9 blood markers. Sci Transl Med. 2017;9:55-83.

41

Walsh AJ, Castellanos JA, Nagathihalli NS, Merchant NB, Skala MC. Optical imaging of drug-induced metabolism changes in murine and human pancreatic cancer organoids reveals heterogeneous drug response. Pancreas. 2016;45:863-869.

42

Li X, Nadauld L, Ootani A, et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20:769-777.

43

Moreira L, Bakir B, Chatterji P, Dantes Z, Reichert M, Rustgi AK. Pancreas 3D organoids: current and future aspects as a research platform for personalized medicine in pancreatic cancer. Cell Mol Gastroenterol Hepatol. 2018;5:289-298.

44

Xu AT, Tong JL, Ran ZH. Organoids derived from digestive tract, liver, and pancreas. J Dig Dis. 2016;17:3-10.

45

Drost J, van Jaarsveld RH, Ponsioen B, et al. Sequential cancer mutations in cultured human intestinal stem cells. Nature. 2015;521:43-47.

46

Matano M, Date S, Shimokawa M, et al. Modeling colorectal cancer using CRISPR-Cas9-mediated engineering of human intestinal organoids. Nat Med. 2015;21:256-262.

47

Fumagalli A, Drost J, Suijkerbuijk SJE, et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc Natl Acad Sci USA. 2017;114:E2357-E2364.

48

Drost J, van Boxtel R, Blokzijl F, et al. Use of CRISPR modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358:234-238.

49

Drost J, Clevers H. Organoids in cancer research. Nat Rev Cancer. 2018;18:407-418.

50

Tsai S, McOlash L, Palen K, et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. 2018;18:335.

51

Finnberg NK, Gokare P, Lev A, et al. Application of 3D tumoroid systems to define immune and cytotoxic therapeutic responses based on tumoroid and tissue slice culture molecular signatures. Oncotarget. 2017;8:66747-66757.

52

Marx V. Cell-line authentication demystified. Nat Methods. 2014;11:483-488.

53

Masters JR, Stacey GN. Changing medium and passaging cell lines. Nat Protoc. 2007;2:2276-2284.

54

Gracz AD. A high-throughput platform for stem cell niche co-cultures and downstream gene expression analysis. Nat Cell Biol. 2015;17:340-349.

55

Cao L. Development of intestinal organoids as tissue surrogates: cell composition and the epigenetic control of differentiation. Mol Carcinog. 2015;54:189-202.

56

Meng Q. Three-dimensional culture of hepatocytes for prediction of drug-induced hepatotoxicity. Expet Opin Drug Metabol Toxicol. 2010;6:733-746.

57

Dekkers JF, Berkers G, Kruisselbrink E, et al. Characterizing responses to CFTRmodulating drugs using rectal organoids derived from subjects with cystic fibrosis. Sci Transl Med. 2016;8:344384.

58

Yui S, Nakamura T, Sato T, et al. Functional engraftment of colon epithelium expanded in vitro from a single adult Lgr5+ stem cell. Nat Med. 2012;18:618-623.

59

Taguchi A, Kaku Y, Ohmori T, Sharmin S, Ogawa M, Sasaki H, Nishinakamura R. Redefining the in vivo origin of metanephric nephron progenitors enables generation of complex kidney structures from pluripotent stem cells. Cell Stem Cell. 2014;14:53-67.

Genes & Diseases
Pages 323-330
Cite this article:
Pan Y, Zhao S, Cao Z. Organoid models of gastrointestinal Neoplasms: Origin, current status and future applications in personalized medicine. Genes & Diseases, 2018, 5(4): 323-330. https://doi.org/10.1016/j.gendis.2018.09.002

314

Views

1

Downloads

8

Crossref

N/A

Web of Science

8

Scopus

0

CSCD

Altmetrics

Received: 07 September 2018
Accepted: 19 September 2018
Published: 24 September 2018
© 2018, Chongqing Medical University.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return