Patient-derived lung cancer organoids as in vitro cancer models for therapeutic screening

Minsuh Kim, Hyemin Mun, Chang Oak Sung, Eun Jeong Cho, Hye-Joon Jeon, Sung-Min Chun, Da Jung Jung, Tae Hoon Shin, Gi Seok Jeong, Dong Kwan Kim, Eun Kyung Choi, Seong-Yun Jeong, Alison M Taylor, Sejal Jain, Matthew Meyerson, Se Jin Jang, Minsuh Kim, Hyemin Mun, Chang Oak Sung, Eun Jeong Cho, Hye-Joon Jeon, Sung-Min Chun, Da Jung Jung, Tae Hoon Shin, Gi Seok Jeong, Dong Kwan Kim, Eun Kyung Choi, Seong-Yun Jeong, Alison M Taylor, Sejal Jain, Matthew Meyerson, Se Jin Jang

Abstract

Lung cancer shows substantial genetic and phenotypic heterogeneity across individuals, driving a need for personalised medicine. Here, we report lung cancer organoids and normal bronchial organoids established from patient tissues comprising five histological subtypes of lung cancer and non-neoplastic bronchial mucosa as in vitro models representing individual patient. The lung cancer organoids recapitulate the tissue architecture of the primary lung tumours and maintain the genomic alterations of the original tumours during long-term expansion in vitro. The normal bronchial organoids maintain cellular components of normal bronchial mucosa. Lung cancer organoids respond to drugs based on their genomic alterations: a BRCA2-mutant organoid to olaparib, an EGFR-mutant organoid to erlotinib, and an EGFR-mutant/MET-amplified organoid to crizotinib. Considering the short length of time from organoid establishment to drug testing, our newly developed model may prove useful for predicting patient-specific drug responses through in vitro patient-specific drug trials.

Conflict of interest statement

The authors declare no competing interests.

Figures

Fig. 1
Fig. 1
LCOs established for the lung cancer biobank. a Bright-field microscopy images of LCOs cultured for 2 weeks. Scale bar, 100 μm. The information of LCOs in these images; LCO-05, LCO-36, and LCO-55; adenocarcinoma, LCO-13; squamous cell carcinoma, LCO-21; small cell carcinoma, LCO-29; large cell carcinoma. b Representative images of long-term cultured LCOs. This LCO was derived from LC-49. Scale bar, 200 μm. c Representative images of successful and failed 2D and 3D cultures derived from lung cancers with different tissue composition. Scale bar, 200 μm. d The graph showing the successful or failed cases according to cancer tissue quality and the establishment rate of each cancer models according to lung cancer subtypes. Cytologic QC, cytologic quality check. e Pie chart showing the subtypes of established 80 LCOs for the lung cancer biobank. The information of 80 LCOs is shown in Table 2. f Bright-field microscopy images and H&E staining images of LCOs before freezing and after thawing. Scale bar, 200 μm. The information of LCOs in these images: LCO-28 — squamous cell carcinoma; LCO-29 — large cell carcinoma; LCO-51 — adenocarcinoma; LCO-75 — small cell carcinoma; LCO-86 — adenosquamous carcinoma
Fig. 2
Fig. 2
LCOs recapitulate the characteristics of the original tissues. ad H&E-stained and IHC-stained images of LCOs and their original LC tissues. The enlarged images in blue boxes of b showed cytoplasmic keratinisation of individual squamous carcinoma cells. Scale bars, 100 μm. e H&E and IHC-stained images of an adenosquamous carcinoma organoid (upper panel). Individual organoids expressed either adenocarcinoma marker (CK7) or squamous cell carcinoma markers (CK5/6 or p63). Some single organoids showed mixed cell pattern composed of CK7+ and CK7− cells, CK5/6+ and CK5/6− cells, and p63+ and p63− cells. Scale bar, 100 μm. Bright field microscopy and immunofluorescence (IF) images of an adenosqumaous carcinoma organoid originated from a single cell (lower panel). The immunofluorescence staining was performed at Day 18 after seeding. The organoid from a single cell was composed of p63+/CK7− cells and p63−/CK7+ cells. The red arrow indicated a single cell seeded in a micro-well. Scale bar in bright-field microscopy images, 100 μm. Scale bar in IF images, 20 μm
Fig. 3
Fig. 3
LCOs retain the genetic characteristics of the original tissues after long-term cultured. a Heat-map analysis of the top 30 mutations in LCOs and their corresponding LC tissues. O LCO, T patient tissue. b Comparison of VAF of genetic alterations detected in LCOs and LC tissues. c Heatmap showing somatic mutations affecting cancer genes in LC tissues and paired LCOs. d Venn diagrams indicating the number of somatic mutations present in each LC tissues and their paired LCOs. e Heatmap showing somatic mutations affecting cancer genes in early passage and late passage organoids. f Venn diagrams indicating the number of somatic mutations present in early passage and late passage organoids
Fig. 4
Fig. 4
Organ-like structures of the NBOs and LCOs. a Bright-field microscopy images of a NBO, NBO-90. NBO-90 was cultured in MBM and MBM + WNA (W; Wnt3A, N; Noggin, A; A83-01). Scale bars, 200 μm. b H&E staining image of NBO-90. Scale bar, 100 μm. ce Immunofluorescence images showing the expression of pancytokeratin, p63, MUC1, KRT5, KRT7, CC10, and cilia co-stained with acetylated α-tubulin (Ac-Tub) and Arl13b (The enlarged images in yellow boxes showed cilia co-stained with Ac-Tub and Arl13b). Nuclei (blue) were stained with DAPI. Scale bar, 20 μm
Fig. 5
Fig. 5
PDXs from LCOs maintain the characteristics of the original tissues. a List of PDXs established by transplanting LCOs or injecting 2D-LC cells. Transplanted LCOs and injected 2D-LC cells that formed tumours in mice are marked as “+”, and the LCOs and 2D-LC cells that did not form tumours are marked as “−”. b Images of mice used to establish PDXs. LCOs (left) and 2D-LC cells (right) were simultaneously transplanted or injected into the same mouse. Circles indicate the formed tumours. c Experimental design and results. Bar graph showing the number of days until tumour formation. n.d: tumour not detected
Fig. 6
Fig. 6
LCOs as a platform for predicting therapeutic responses. a Dose-response curves after 6 days of treatment with docetaxel. The cell viability was measured by luminescence signal intensities. b and c Dose-response curves after 6 days of treatment with olaparib b or erlotinib c. The indicated drug concentrations were used to treat two LCO lines with mutations (green box), another two LCO lines without mutations (grey box) and the NBO. Representative viability curves were generated from luminescence signal intensities. d The copy number variation (CNV) graph showing MET copy number amplification in the four LCO lines. e IHC-stained images showing c-MET expression in the four patient tissues and LCOs. Scale bar, 100 μm. f Dose-response curves after treatment with indicated drug concentrations of crizotinib in four LCOs and a NBO. Representative viability curves were generated from the luminescence signal intensities. g Immunoblot analysis showing changed protein expressions mediating EGFR signalling and c-Met signalling transduction after treating erlotinib and crizotinib. Two LCOs, LCO-43 and LCO-51 were treated with 1 μM erlotinib and 1 μM crizotinib for 24, 48 and 72 h. GAPDH was used as a loading control. Full-length blot is shown in the supplementary information, and the gels have been run under the same experimental conditions. All error bars in ac and f indicate SEM, n = 3. In b, c and f IC50 values are the average ± SD of each condition analysed in triplicate. When error bars are not visible they are smaller than the size of the symbol

References

    1. Li S, et al. Enhanced recovery programs in lung cancer surgery: systematic review and meta-analysis of randomized controlled trials. Cancer Manag. Res. 2017;9:657–670. doi: 10.2147/CMAR.S150500.
    1. Herbst. RS, Heymach JV, Lippman SM. Lung cancer. N. Engl. J. Med. 2008;359:1367–1380. doi: 10.1056/NEJMra0802714.
    1. Reck M, et al. Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N. Engl. J. Med. 2016;375:1823–1833. doi: 10.1056/NEJMoa1606774.
    1. Sachs N, Clevers H. Organoid cultures for the analysis of cancer phenotypes. Curr. Opin. Genet. Dev. 2014;24:68–73. doi: 10.1016/j.gde.2013.11.012.
    1. Chen Z, Fillmore CM, Hammerman PS, Kim CF, Wong KK. Non-small-cell lung cancers: a heterogeneous set of diseases. Nat. Rev. Cancer. 2014;14:535–546. doi: 10.1038/nrc3775.
    1. Siolas D, Hannon GJ. Patient-derived tumor xenografts: transforming clinical samples into mouse models. Cancer Res. 2013;73:5315–5319. doi: 10.1158/0008-5472.CAN-13-1069.
    1. Tentler JJ, et al. Patient-derived tumour xenografts as models for oncology drug development. Nat. Rev. Clin. Oncol. 2012;9:338–350. doi: 10.1038/nrclinonc.2012.61.
    1. Guenot D, et al. Primary tumour genetic alterations and intra-tumoral heterogeneity are maintained in xenografts of human colon cancers showing chromosome instability. J. Pathol. 2006;208:643–652. doi: 10.1002/path.1936.
    1. Dong X, et al. Patient-derived first generation xenografts of non-small cell lung cancers: promising tools for predicting drug responses for personalized chemotherapy. Clin. Cancer Res. 2010;16:1442–1451. doi: 10.1158/1078-0432.CCR-09-2878.
    1. Zhang Z, et al. Establishment of patient-derived tumor spheroids for non-small cell lung cancer. PLoS One. 2018;13:e0194016. doi: 10.1371/journal.pone.0194016.
    1. Sato T, et al. Long-term expansion of epithelial organoids from human colon, adenoma, adenocarcinoma, and Barrett’s epithelium. Gastroenterology. 2011;141:1762–1772. doi: 10.1053/j.gastro.2011.07.050.
    1. Bartfeld S, et al. In vitro expansion of human gastric epithelial stem cells and their responses to bacterial infection. Gastroenterology. 2015;148:126–136.e126. doi: 10.1053/j.gastro.2014.09.042.
    1. Broutier L, et al. Culture and establishment of self-renewing human and mouse adult liver and pancreas 3D organoids and their genetic manipulation. Nat. Protoc. 2016;11:1724–1743. doi: 10.1038/nprot.2016.097.
    1. Barkauskas CE, et al. Lung organoids: current uses and future promise. Development. 2017;144:986–997. doi: 10.1242/dev.140103.
    1. Sasai Y, Eiraku M, Suga H. In vitro organogenesis in three dimensions: self-organising stem cells. Development. 2012;139:4111–4121. doi: 10.1242/dev.079590.
    1. Pauli C, et al. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov. 2017;7:462–477. doi: 10.1158/-16-1154.
    1. Shamir ER, Ewald AJ. Three-dimensional organotypic culture: experimental models of mammalian biology and disease. Nat. Rev. Mol. Cell Biol. 2014;15:647–664. doi: 10.1038/nrm3873.
    1. Van de Wetering M, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015;161:933–945. doi: 10.1016/j.cell.2015.03.053.
    1. Boj SF, et al. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015;160:324–338. doi: 10.1016/j.cell.2014.12.021.
    1. Gao D, et al. Organoid cultures derived from patients with advanced prostate cancer. Cell. 2014;159:176–187. doi: 10.1016/j.cell.2014.08.016.
    1. Broutier L, et al. Human primary liver cancer-derived organoid cultures for disease modeling and drug screening. Nat. Med. 2017;23:1424–1435. doi: 10.1038/nm.4438.
    1. Sachs N, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018;172:373–386 e310. doi: 10.1016/j.cell.2017.11.010.
    1. Drost J, Clevers H. Organoids in cancer research. Nat. Rev. Cancer. 2018;18:407–418. doi: 10.1038/s41568-018-0007-6.
    1. Vlachogiannis G, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018;359:920–926. doi: 10.1126/science.aao2774.
    1. Choi J, Iich E, Lee JH. Organogenesis of adult lung in a dish: Differentiation, disease and therapy. Dev. Biol. 2016;420:278–286. doi: 10.1016/j.ydbio.2016.10.002.
    1. Dye, B. et al. In vitro generation of human pluripotent stem cell derived lung organoids. eLife10.7554/eLife.05098 (2015).
    1. Karekla E, et al. Ex vivo explant cultures of non-small cell lung carcinoma enable evaluation of primary tumor responses to anticancer therapy. Cancer Res. 2017;77:2029–2039. doi: 10.1158/0008-5472.CAN-16-1121.
    1. Chen YW, et al. A three-dimensional model of human lung development and disease from pluripotent stem cells. Nat. Cell Biol. 2017;19:542–549. doi: 10.1038/ncb3510.
    1. McCauley KB, et al. Efficient derivation of functional human airway epithelium from pluripotent stem cells via temporal regulation of Wnt signaling. Cell Stem Cell. 2017;20:844–857. doi: 10.1016/j.stem.2017.03.001.
    1. Jenkins RW, et al. Ex vivo profiling of PD-1 blockade using organotypic tumor spheroids. Cancer Discov. 2018;8:196–215. doi: 10.1158/-17-0833.
    1. Sachs, N. et al. Long-term expanding human airway organoids for disease modeling. EMBO J. 38, e100300 (2019).
    1. Zhang WC, et al. Glycine decarboxylase activity drives non-small cell lung cancer tumor-initiating cells and tumorigenesis. Cell. 2012;148:259–272. doi: 10.1016/j.cell.2011.11.050.
    1. Chen WJ, et al. Cancer-associated fibroblasts regulate the plasticity of lung cancer stemness via paracrine signalling. Nat. Commun. 2014;5:3472. doi: 10.1038/ncomms4472.
    1. Fasano M, Della Corte CM, Papaccio F, Ciardiello F, Morgillo F. Pulmonary large-cell neuroendocrine carcinoma: from epidemiology to therapy. J. Thorac. Oncol. 2015;10:1133–1141. doi: 10.1097/JTO.0000000000000589.
    1. Crabtree JS, Miele L. Neuroendocrine tumors: current therapies, notch signaling, and cancer stem cells. J. Cancer Metastasis Treat. 2016;2:279. doi: 10.20517/2394-4722.2016.30.
    1. Mia-Jan K, et al. Expression of CD133 in neuroendocrine neoplasms of the digestive tract: a detailed immunohistochemical analysis. Tohoku J. Exp. Med. 2013;229:301–309. doi: 10.1620/tjem.229.301.
    1. Thunnissen E, et al. The use of immunohistochemistry improves the diagnosis of small cell lung cancer and its differential diagnosis. an international reproducibility study in a demanding set of cases. J. Thorac. Oncol. 2017;12:334–346. doi: 10.1016/j.jtho.2016.12.004.
    1. Nuciforo S, et al. Organoid models of human liver cancers derived from tumor needle biopsies. Cell Rep. 2018;24:1363–1376. doi: 10.1016/j.celrep.2018.07.001.
    1. Shen S, et al. Mutant-allele fraction heterogeneity is associated with non-small cell lung cancer patient survival. Oncol. Lett. 2018;15:795–802.
    1. Dienstmann R, et al. Analysis of mutant allele fractions in driver genes in colorectal cancer—biological and clinical insights. Mol. Oncol. 2017;11:1263–1272. doi: 10.1002/1878-0261.12099.
    1. Reynolds A, et al. Canonical Wnt signals combined with suppressed TGFbeta/BMP pathways promote renewal of the native human colonic epithelium. Gut. 2014;63:610–621. doi: 10.1136/gutjnl-2012-304067.
    1. Nakajima M, et al. Immunohistochemical and ultrastructural studies of basal cells, Clara cells and bronchiolar cuboidal cells in normal human airways. Pathol. Int. 1998;48:944–953. doi: 10.1111/j.1440-1827.1998.tb03865.x.
    1. Hiemstra PS, Bourdin A. Club cells, CC10 and self-control at the epithelial surface. Eur. Respir. J. 2014;44:831–832. doi: 10.1183/09031936.00089214.
    1. Kim S, Dynlacht BD. Assembling a primary cilium. Curr. Opin. Cell Biol. 2013;25:506–511. doi: 10.1016/j.ceb.2013.04.011.
    1. Huang L, 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. doi: 10.1038/nm.3973.
    1. Farkona S, Diamandis EP, Blasutig IM. Cancer immunotherapy: the beginning of the end of cancer? BMC Med. 2016;14:73. doi: 10.1186/s12916-016-0623-5.
    1. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–674. doi: 10.1016/j.cell.2011.02.013.
    1. Seetharamu N, Preeshagul IR, Sullivan KM. New PD-L1 inhibitors in non-small cell lung cancer - impact of atezolizumab. Lung Cancer. 2017;8:67–78.
    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 2015;372:2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Li Yunfeng, Kumacheva Eugenia. Hydrogel microenvironments for cancer spheroid growth and drug screening. Science Advances. 2018;4(4):eaas8998. doi: 10.1126/sciadv.aas8998.
    1. Czyż M, et al. Enhancing synthetic lethality of PARP-inhibitor and cisplatin in BRCA-proficient tumour cells with hyperthermia. Oncotarget. 2016;7:75551–75560. doi: 10.18632/oncotarget.12270.
    1. Helleday T. The underlying mechanism for the PARP and BRCA synthetic lethality: clearing up the misunderstandings. Mol. Oncol. 2011;5:387–393. doi: 10.1016/j.molonc.2011.07.001.
    1. Dziadkowiec KN, Gasiorowska E, Nowak-Markwitz E, Jankowska A. PARP inhibitors: review of mechanisms of action and BRCA1/2 mutation targeting. Prz. Menopauzalny. 2016;15:215–219.
    1. Adzhubei, I., Jordan, D. M., Sunyaev, S. R. Predicting functional effect of human missense mutations using PolyPhen-2. Curr. Protoc. Hum. Genet.7, 7.20 (2013).
    1. Costa DB, et al. Effects of erlotinib in EGFR mutated non-small cell lung cancers with resistance to gefitinib. Clin. Cancer Res. 2008;14:7060–7067. doi: 10.1158/1078-0432.CCR-08-1455.
    1. LL S, L L, WH Z, WD H. Establishment and identification of human primary lung cancer cell culture in vitro. Int. J. Clin. Exp. Pathol. 2015;8:6540–6546.
    1. J. Z, et al. Sensitivity of neoplastic cells to senescence unveiled under standard cell culture conditions. Anticancer Res. 2015;35:2759–2768.
    1. Nikolic MZ, Rawlins EL. Lung organoids and their use to study cell–cell interaction. Curr. Pathobiol. Rep. 2017;5:223–231. doi: 10.1007/s40139-017-0137-7.
    1. Weeber F, Ooft SN, Dijkstra KK, Voest EE. Tumor organoids as a pre-clinical cancer model for drug discovery. Cell Chem. Biol. 2017;24:1092–1100. doi: 10.1016/j.chembiol.2017.06.012.
    1. Palucka AK, Coussens LM. The basis of oncoimmunology. Cell. 2016;164:1233–1247. doi: 10.1016/j.cell.2016.01.049.
    1. Dudvarski Stankovic, N. et al. EGFL7 enhances surface expression of integrin alpha5beta1 to promote angiogenesis in malignant brain tumors. EMBO Mol. Med.10, e8420 (2018).
    1. Pikor LA, Ramnarine VR, Lam S, Lam WL. Genetic alterations defining NSCLC subtypes and their therapeutic implications. Lung Cancer. 2013;82:179–189. doi: 10.1016/j.lungcan.2013.07.025.
    1. Boyd. JA, et al. Timing of local and distant failure in resected lung cancer implications for reported rates of local failure. J. Thorac. Oncol. 2010;5:211–214. doi: 10.1097/JTO.0b013e3181c20080.
    1. McKenna A, et al. The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data. Genome Res. 2010;20:1297–1303. doi: 10.1101/gr.107524.110.
    1. Li H, Durbin R. Fast and accurate long-read alignment with Burrows–Wheeler transform. Bioinformatics. 2010;26:589–595. doi: 10.1093/bioinformatics/btp698.
    1. DePristo MA, et al. A framework for variation discovery and genotyping using next-generation DNA sequencing data. Nat. Genet. 2011;43:491–498. doi: 10.1038/ng.806.
    1. Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat. Biotechnol. 2013;31:213–219. doi: 10.1038/nbt.2514.
    1. Talevich E, Shain AH, Botton T, Bastian BC. CNVkit: genome-wide copy number detection and visualization from targeted DNA sequencing. PLoS Comput. Biol. 2016;12:e1004873. doi: 10.1371/journal.pcbi.1004873.
    1. Robinson J, et al. Integrative genomics viewer. Nat. Biotechnol. 2011;29:24–26. doi: 10.1038/nbt.1754.

Source: PubMed

3
订阅