Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2019

Open Access 01-12-2019 | Metastasis | Review

Emerging organoid models: leaping forward in cancer research

Authors: Han Fan, Utkan Demirci, Pu Chen

Published in: Journal of Hematology & Oncology | Issue 1/2019

Login to get access

Abstract

Cancer heterogeneity is regarded as the main reason for the failure of conventional cancer therapy. The ability to reconstruct intra- and interpatient heterogeneity in cancer models is crucial for understanding cancer biology as well as for developing personalized anti-cancer therapy. Cancer organoids represent an emerging approach for creating patient-derived in vitro cancer models that closely recapitulate the pathophysiological features of natural tumorigenesis and metastasis. Meanwhile, cancer organoids have recently been utilized in the discovery of personalized anti-cancer therapy and prognostic biomarkers. Further, the synergistic combination of cancer organoids with organ-on-a-chip and 3D bioprinting presents a new avenue in the development of more sophisticated and optimized model systems to recapitulate complex cancer-stroma or multiorgan metastasis. Here, we summarize the recent advances in cancer organoids from a perspective of the in vitro emulation of natural cancer evolution and the applications in personalized cancer theranostics. We also discuss the challenges and trends in reconstructing more comprehensive cancer models for basic and clinical cancer research.
Literature
1.
go back to reference Society AC. Global Cancer Facts & Figures 3rd Edition. Am Cancer Soc. 2015;800:1–64. Society AC. Global Cancer Facts & Figures 3rd Edition. Am Cancer Soc. 2015;800:1–64.
2.
go back to reference McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613–28.PubMedCrossRef McGranahan N, Swanton C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell. 2017;168(4):613–28.PubMedCrossRef
4.
go back to reference Torsvik A, Stieber D, Enger PØ, Golebiewska A, Molven A, Svendsen A, et al. U-251 revisited: genetic drift and phenotypic consequences of long-term cultures of glioblastoma cells. Cancer medicine. 2014;3(4):812–24.PubMedPubMedCentralCrossRef Torsvik A, Stieber D, Enger PØ, Golebiewska A, Molven A, Svendsen A, et al. U-251 revisited: genetic drift and phenotypic consequences of long-term cultures of glioblastoma cells. Cancer medicine. 2014;3(4):812–24.PubMedPubMedCentralCrossRef
5.
go back to reference Li S, Shen D, Shao J, Crowder R, Liu W, Prat A, et al. Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts. Cell Rep. 2013;4(6):1116–30.PubMedCrossRef Li S, Shen D, Shao J, Crowder R, Liu W, Prat A, et al. Endocrine-therapy-resistant ESR1 variants revealed by genomic characterization of breast-cancer-derived xenografts. Cell Rep. 2013;4(6):1116–30.PubMedCrossRef
7.
8.
go back to reference Pergolini I, Morales-Oyarvide V, Mino-Kenudson M, et al. Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival. PLoS One. 2017;12(8):e0182855.PubMedPubMedCentralCrossRef Pergolini I, Morales-Oyarvide V, Mino-Kenudson M, et al. Tumor engraftment in patient-derived xenografts of pancreatic ductal adenocarcinoma is associated with adverse clinicopathological features and poor survival. PLoS One. 2017;12(8):e0182855.PubMedPubMedCentralCrossRef
9.
go back to reference van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.PubMedPubMedCentralCrossRef van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161(4):933–45.PubMedPubMedCentralCrossRef
10.
go back to reference Sachs N, de Ligt J, Kopper O, et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018;172(1-2):373–386.e10.PubMedCrossRef Sachs N, de Ligt J, Kopper O, et al. A Living Biobank of Breast Cancer Organoids Captures Disease Heterogeneity. Cell. 2018;172(1-2):373–386.e10.PubMedCrossRef
11.
go back to reference Nuciforo S, Fofana I, Matter MS, Blumer T, Calabrese D, Boldanova T, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24(5):1363–76.PubMedPubMedCentralCrossRef Nuciforo S, Fofana I, Matter MS, Blumer T, Calabrese D, Boldanova T, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24(5):1363–76.PubMedPubMedCentralCrossRef
12.
go back to reference Saito Y, Muramatsu T, Kanai Y, et al. Establishment of Patient-Derived Organoids and Drug Screening for Biliary Tract Carcinoma. Cell Rep. 2019;27(4):1265–1276.e4.PubMedCrossRef Saito Y, Muramatsu T, Kanai Y, et al. Establishment of Patient-Derived Organoids and Drug Screening for Biliary Tract Carcinoma. Cell Rep. 2019;27(4):1265–1276.e4.PubMedCrossRef
13.
go back to reference Dijkstra KK, Cattaneo CM, Weeber F, et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 2018;174(6):1586–1598.e12.PubMedPubMedCentralCrossRef Dijkstra KK, Cattaneo CM, Weeber F, et al. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 2018;174(6):1586–1598.e12.PubMedPubMedCentralCrossRef
14.
go back to reference 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(40):66747–57.PubMedPubMedCentralCrossRef 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(40):66747–57.PubMedPubMedCentralCrossRef
16.
go back to reference Della Corte CM, Barra G, Ciaramella V, et al. Antitumor activity of dual blockade of PD-L1 and MEK in NSCLC patients derived three-dimensional spheroid cultures. J Exp Clin Cancer Res. 2019;38(1):253.PubMedPubMedCentralCrossRef Della Corte CM, Barra G, Ciaramella V, et al. Antitumor activity of dual blockade of PD-L1 and MEK in NSCLC patients derived three-dimensional spheroid cultures. J Exp Clin Cancer Res. 2019;38(1):253.PubMedPubMedCentralCrossRef
18.
19.
go back to reference Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer discovery. 2014;4(9):998–1013.PubMedPubMedCentralCrossRef Hidalgo M, Amant F, Biankin AV, Budinská E, Byrne AT, Caldas C, et al. Patient-derived xenograft models: an emerging platform for translational cancer research. Cancer discovery. 2014;4(9):998–1013.PubMedPubMedCentralCrossRef
20.
go back to reference Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol. 2016;18(3):246.PubMedCrossRef Fatehullah A, Tan SH, Barker N. Organoids as an in vitro model of human development and disease. Nat Cell Biol. 2016;18(3):246.PubMedCrossRef
23.
go back to reference Walsh AJ, Cook RS, Sanders ME, Aurisicchio L, Ciliberto G, Arteaga CL, et al. Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. 2014;74(18):5184–94.PubMedPubMedCentralCrossRef Walsh AJ, Cook RS, Sanders ME, Aurisicchio L, Ciliberto G, Arteaga CL, et al. Quantitative optical imaging of primary tumor organoid metabolism predicts drug response in breast cancer. Cancer Res. 2014;74(18):5184–94.PubMedPubMedCentralCrossRef
24.
go back to reference Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature. 2016;540(7634):588.PubMedPubMedCentralCrossRef Harper KL, Sosa MS, Entenberg D, Hosseini H, Cheung JF, Nobre R, et al. Mechanism of early dissemination and metastasis in Her2+ mammary cancer. Nature. 2016;540(7634):588.PubMedPubMedCentralCrossRef
25.
go back to reference Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 2014;46(6):573.PubMedCrossRef Wang K, Yuen ST, Xu J, Lee SP, Yan HH, Shi ST, et al. Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet. 2014;46(6):573.PubMedCrossRef
26.
go back to reference Nadauld LD, Garcia S, Natsoulis G, Bell JM, Miotke L, Hopmans ES, et al. Metastatic tumor evolution and organoid modeling implicate TGFBR2 as a cancer driver in diffuse gastric cancer. Genome Biol. 2014;15(8):428.PubMedPubMedCentralCrossRef Nadauld LD, Garcia S, Natsoulis G, Bell JM, Miotke L, Hopmans ES, et al. Metastatic tumor evolution and organoid modeling implicate TGFBR2 as a cancer driver in diffuse gastric cancer. Genome Biol. 2014;15(8):428.PubMedPubMedCentralCrossRef
27.
go back to reference Drost J, Van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358(6360):234–8.PubMedPubMedCentralCrossRef Drost J, Van Boxtel R, Blokzijl F, Mizutani T, Sasaki N, Sasselli V, et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science. 2017;358(6360):234–8.PubMedPubMedCentralCrossRef
28.
go back to reference Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarró LM, Bradshaw CR, et al. Human primary liver cancer–derived organoid cultures for disease modeling and drug screening. Nat Med. 2017;23(12):1424.PubMedPubMedCentralCrossRef Broutier L, Mastrogiovanni G, Verstegen MM, Francies HE, Gavarró LM, Bradshaw CR, et al. Human primary liver cancer–derived organoid cultures for disease modeling and drug screening. Nat Med. 2017;23(12):1424.PubMedPubMedCentralCrossRef
29.
go back to reference Boj SF, Hwang C-I, Baker LA, Chio IIC, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1-2):324–38.PubMedCrossRef Boj SF, Hwang C-I, Baker LA, Chio IIC, Engle DD, Corbo V, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160(1-2):324–38.PubMedCrossRef
30.
go back to reference Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176–87.PubMedPubMedCentralCrossRef Gao D, Vela I, Sboner A, Iaquinta PJ, Karthaus WR, Gopalan A, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159(1):176–87.PubMedPubMedCentralCrossRef
31.
go back to reference Tiriac H, Belleau P, Engle DD, Plenker D, Deschênes A, Somerville TD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer discovery. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef Tiriac H, Belleau P, Engle DD, Plenker D, Deschênes A, Somerville TD, et al. Organoid profiling identifies common responders to chemotherapy in pancreatic cancer. Cancer discovery. 2018;8(9):1112–29.PubMedPubMedCentralCrossRef
32.
go back to reference Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. 2018;18(1):335.PubMedPubMedCentralCrossRef Tsai S, McOlash L, Palen K, Johnson B, Duris C, Yang Q, et al. Development of primary human pancreatic cancer organoids, matched stromal and immune cells and 3D tumor microenvironment models. BMC Cancer. 2018;18(1):335.PubMedPubMedCentralCrossRef
33.
go back to reference Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC, et al. A three dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer Res. 2016;76(8):2465–77.PubMedPubMedCentralCrossRef Hubert CG, Rivera M, Spangler LC, Wu Q, Mack SC, Prager BC, et al. A three dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo. Cancer Res. 2016;76(8):2465–77.PubMedPubMedCentralCrossRef
34.
go back to reference Fumagalli A, Drost J, Suijkerbuijk SJ, Van Boxtel R, De Ligt J, Offerhaus GJ, et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc Natl Acad Sci. 2017;114(12):E2357–E64.PubMedCrossRefPubMedCentral Fumagalli A, Drost J, Suijkerbuijk SJ, Van Boxtel R, De Ligt J, Offerhaus GJ, et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc Natl Acad Sci. 2017;114(12):E2357–E64.PubMedCrossRefPubMedCentral
35.
36.
go back to reference Thoma CR, Zimmermann M, Agarkova I, Kelm JM, Krek W. 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv Drug Deliv Rev. 2014;69:29–41.PubMedCrossRef Thoma CR, Zimmermann M, Agarkova I, Kelm JM, Krek W. 3D cell culture systems modeling tumor growth determinants in cancer target discovery. Adv Drug Deliv Rev. 2014;69:29–41.PubMedCrossRef
37.
go back to reference Onuma K, Ochiai M, Orihashi K, Takahashi M, Imai T, Nakagama H, et al. Genetic reconstitution of tumorigenesis in primary intestinal cells. Proc Natl Acad Sci. 2013;110(27):11127–32.PubMedCrossRefPubMedCentral Onuma K, Ochiai M, Orihashi K, Takahashi M, Imai T, Nakagama H, et al. Genetic reconstitution of tumorigenesis in primary intestinal cells. Proc Natl Acad Sci. 2013;110(27):11127–32.PubMedCrossRefPubMedCentral
38.
go back to reference Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256.PubMedCrossRef Matano M, Date S, Shimokawa M, Takano A, Fujii M, Ohta Y, et al. Modeling colorectal cancer using CRISPR-Cas9–mediated engineering of human intestinal organoids. Nat Med. 2015;21(3):256.PubMedCrossRef
39.
go back to reference Nakayama M, Sakai E, Echizen K, Yamada Y, Oshima H, Han T, et al. Intestinal cancer progression by mutant p53 through the acquisition of invasiveness associated with complex glandular formation. Oncogene. 2017;36(42):5885.PubMedPubMedCentralCrossRef Nakayama M, Sakai E, Echizen K, Yamada Y, Oshima H, Han T, et al. Intestinal cancer progression by mutant p53 through the acquisition of invasiveness associated with complex glandular formation. Oncogene. 2017;36(42):5885.PubMedPubMedCentralCrossRef
40.
go back to reference Schell MJ, Yang M, Teer JK, Lo FY, Madan A, Coppola D, et al. A multigene mutation classification of 468 colorectal cancers reveals a prognostic role for APC. Nat Commun. 2016;7:11743.PubMedPubMedCentralCrossRef Schell MJ, Yang M, Teer JK, Lo FY, Madan A, Coppola D, et al. A multigene mutation classification of 468 colorectal cancers reveals a prognostic role for APC. Nat Commun. 2016;7:11743.PubMedPubMedCentralCrossRef
41.
go back to reference Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20(7):769.PubMedPubMedCentralCrossRef Li X, Nadauld L, Ootani A, Corney DC, Pai RK, Gevaert O, et al. Oncogenic transformation of diverse gastrointestinal tissues in primary organoid culture. Nat Med. 2014;20(7):769.PubMedPubMedCentralCrossRef
42.
go back to reference Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef Fujii M, Shimokawa M, Date S, Takano A, Matano M, Nanki K, et al. A colorectal tumor organoid library demonstrates progressive loss of niche factor requirements during tumorigenesis. Cell Stem Cell. 2016;18(6):827–38.PubMedCrossRef
43.
44.
go back to reference Friedl P, Locker J, Sahai E, Segall JE. Classifying collective cancer cell invasion. Nat Cell Biol. 2012;14(8):777.PubMedCrossRef Friedl P, Locker J, Sahai E, Segall JE. Classifying collective cancer cell invasion. Nat Cell Biol. 2012;14(8):777.PubMedCrossRef
45.
go back to reference Cheung KJ, Gabrielson E, Werb Z, Ewald AJ. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell. 2013;155(7):1639–51.PubMedPubMedCentralCrossRef Cheung KJ, Gabrielson E, Werb Z, Ewald AJ. Collective invasion in breast cancer requires a conserved basal epithelial program. Cell. 2013;155(7):1639–51.PubMedPubMedCentralCrossRef
46.
go back to reference Wu JS, Li ZF, Wang HF, et al. Cathepsin B defines leader cells during the collective invasion of salivary adenoid cystic carcinoma. Int J Oncol. 2019;54(4):1233–44.PubMedPubMedCentral Wu JS, Li ZF, Wang HF, et al. Cathepsin B defines leader cells during the collective invasion of salivary adenoid cystic carcinoma. Int J Oncol. 2019;54(4):1233–44.PubMedPubMedCentral
47.
48.
go back to reference Björk JK, Åkerfelt M, Joutsen J, et al. Heat-shock factor 2 is a suppressor of prostate cancer invasion. Oncogene. 2016;35(14):1770–84.PubMedCrossRef Björk JK, Åkerfelt M, Joutsen J, et al. Heat-shock factor 2 is a suppressor of prostate cancer invasion. Oncogene. 2016;35(14):1770–84.PubMedCrossRef
49.
go back to reference Vellinga TT, den Uil S, Rinkes IH, et al. Collagen-rich stroma in aggressive colon tumors induces mesenchymal gene expression and tumor cell invasion. Oncogene. 2016;35(40):5263–71.PubMedCrossRef Vellinga TT, den Uil S, Rinkes IH, et al. Collagen-rich stroma in aggressive colon tumors induces mesenchymal gene expression and tumor cell invasion. Oncogene. 2016;35(40):5263–71.PubMedCrossRef
50.
go back to reference Chandhoke AS, Chanda A, Karve K, Deng L, Bonni S. The PIAS3-Smurf2 sumoylation pathway suppresses breast cancer organoid invasiveness. Oncotarget. 2017;8(13):21001. Chandhoke AS, Chanda A, Karve K, Deng L, Bonni S. The PIAS3-Smurf2 sumoylation pathway suppresses breast cancer organoid invasiveness. Oncotarget. 2017;8(13):21001.
51.
go back to reference Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833(12):3481–98.PubMedCrossRef Paoli P, Giannoni E, Chiarugi P. Anoikis molecular pathways and its role in cancer progression. Biochim Biophys Acta. 2013;1833(12):3481–98.PubMedCrossRef
53.
go back to reference Hattar R, Maller O, McDaniel S, Hansen KC, Hedman KJ, Lyons TR, et al. Tamoxifen induces pleiotrophic changes in mammary stroma resulting in extracellular matrix that suppresses transformed phenotypes. Breast Cancer Res. 2009;11(1):R5.PubMedPubMedCentralCrossRef Hattar R, Maller O, McDaniel S, Hansen KC, Hedman KJ, Lyons TR, et al. Tamoxifen induces pleiotrophic changes in mammary stroma resulting in extracellular matrix that suppresses transformed phenotypes. Breast Cancer Res. 2009;11(1):R5.PubMedPubMedCentralCrossRef
54.
go back to reference Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, et al. Fibronectin rescues estrogen receptor α from lysosomal degradation in breast cancer cells. J Cell Biol. 2018;217(8):2777–98.PubMedPubMedCentralCrossRef Sampayo RG, Toscani AM, Rubashkin MG, Thi K, Masullo LA, Violi IL, et al. Fibronectin rescues estrogen receptor α from lysosomal degradation in breast cancer cells. J Cell Biol. 2018;217(8):2777–98.PubMedPubMedCentralCrossRef
55.
go back to reference Gao H, Chakraborty G, Zhang Z, Akalay I, Gadiya M, Gao Y, et al. Multi-organ site metastatic reactivation mediated by non-canonical discoidin domain receptor 1 signaling. Cell. 2016;166(1):47–62.PubMedPubMedCentralCrossRef Gao H, Chakraborty G, Zhang Z, Akalay I, Gadiya M, Gao Y, et al. Multi-organ site metastatic reactivation mediated by non-canonical discoidin domain receptor 1 signaling. Cell. 2016;166(1):47–62.PubMedPubMedCentralCrossRef
56.
go back to reference Wilding JL, Bodmer WF. Cancer cell lines for drug discovery and development. Cancer Res. 2014;74(9):2377–84.PubMedCrossRef Wilding JL, Bodmer WF. Cancer cell lines for drug discovery and development. Cancer Res. 2014;74(9):2377–84.PubMedCrossRef
57.
go back to reference Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32(1):40.PubMedCrossRef Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32(1):40.PubMedCrossRef
59.
go back to reference Sahin U. Studying tumor-reactive t cells: a personalized organoid model. Cell Stem Cell. 2018;23(3):318–9.PubMedCrossRef Sahin U. Studying tumor-reactive t cells: a personalized organoid model. Cell Stem Cell. 2018;23(3):318–9.PubMedCrossRef
60.
62.
go back to reference Vlachogiannis G, Hedayat S, Vatsiou A, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.PubMedPubMedCentralCrossRef Vlachogiannis G, Hedayat S, Vatsiou A, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359(6378):920–6.PubMedPubMedCentralCrossRef
64.
go back to reference Takebe T, Zhang B, Radisic M. Synergistic engineering: organoids meet organs-on-a-chip. Cell Stem Cell. 2017;21(3):297–300.PubMedCrossRef Takebe T, Zhang B, Radisic M. Synergistic engineering: organoids meet organs-on-a-chip. Cell Stem Cell. 2017;21(3):297–300.PubMedCrossRef
65.
go back to reference Jeon JS, Bersini S, Gilardi M, Dubini G, Charest JL, Moretti M, et al. Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation. Proc Natl Acad Sci. 2015;112(1):214–9.PubMedCrossRef Jeon JS, Bersini S, Gilardi M, Dubini G, Charest JL, Moretti M, et al. Human 3D vascularized organotypic microfluidic assays to study breast cancer cell extravasation. Proc Natl Acad Sci. 2015;112(1):214–9.PubMedCrossRef
66.
go back to reference Xu Z, Li E, Guo Z, Yu R, Hao H, Xu Y, et al. Design and construction of a multi-organ microfluidic chip mimicking the in vivo microenvironment of lung cancer metastasis. ACS Appl Mater Interfaces. 2016;8(39):25840–7.PubMedCrossRef Xu Z, Li E, Guo Z, Yu R, Hao H, Xu Y, et al. Design and construction of a multi-organ microfluidic chip mimicking the in vivo microenvironment of lung cancer metastasis. ACS Appl Mater Interfaces. 2016;8(39):25840–7.PubMedCrossRef
67.
go back to reference Peng W, Unutmaz D, Ozbolat IT. Bioprinting towards physiologically relevant tissue models for pharmaceutics. Trends Biotechnol. 2016;34(9):722–32.PubMedCrossRef Peng W, Unutmaz D, Ozbolat IT. Bioprinting towards physiologically relevant tissue models for pharmaceutics. Trends Biotechnol. 2016;34(9):722–32.PubMedCrossRef
68.
go back to reference Lee H, Cho D-W. One-step fabrication of an organ-on-a-chip with spatial heterogeneity using a 3D bioprinting technology. Lab Chip. 2016;16(14):2618–25.PubMedCrossRef Lee H, Cho D-W. One-step fabrication of an organ-on-a-chip with spatial heterogeneity using a 3D bioprinting technology. Lab Chip. 2016;16(14):2618–25.PubMedCrossRef
69.
go back to reference Knowlton S, Yenilmez B, Tasoglu S. Towards single-step biofabrication of organs on a chip via 3D printing. Trends Biotechnol. 2016;34(9):685–8.PubMedCrossRef Knowlton S, Yenilmez B, Tasoglu S. Towards single-step biofabrication of organs on a chip via 3D printing. Trends Biotechnol. 2016;34(9):685–8.PubMedCrossRef
70.
go back to reference Ho CMB, Ng SH, Li KHH, Yoon Y-J. 3D printed microfluidics for biological applications. Lab Chip. 2015;15(18):3627–37.PubMedCrossRef Ho CMB, Ng SH, Li KHH, Yoon Y-J. 3D printed microfluidics for biological applications. Lab Chip. 2015;15(18):3627–37.PubMedCrossRef
71.
72.
go back to reference Pang Y, Mao SS, Yao R, et al. TGF-β induced epithelial-mesenchymal transition in an advanced cervical tumor model by 3D printing. Biofabrication. 2018;10(4):044102.PubMedCrossRef Pang Y, Mao SS, Yao R, et al. TGF-β induced epithelial-mesenchymal transition in an advanced cervical tumor model by 3D printing. Biofabrication. 2018;10(4):044102.PubMedCrossRef
Metadata
Title
Emerging organoid models: leaping forward in cancer research
Authors
Han Fan
Utkan Demirci
Pu Chen
Publication date
01-12-2019
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2019
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-019-0832-4

Other articles of this Issue 1/2019

Journal of Hematology & Oncology 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine