Three-Dimensional Patient-Derived In Vitro Sarcoma Models: Promising Tools for Improving Clinical Tumor Management

Manuela Gaebler, Alessandra Silvestri, Johannes Haybaeck, Peter Reichardt, Caitlin D Lowery, Louis F Stancato, Gabriele Zybarth, Christian R A Regenbrecht, Manuela Gaebler, Alessandra Silvestri, Johannes Haybaeck, Peter Reichardt, Caitlin D Lowery, Louis F Stancato, Gabriele Zybarth, Christian R A Regenbrecht

Abstract

Over the past decade, the development of new targeted therapeutics directed against specific molecular pathways involved in tumor cell proliferation and survival has allowed an essential improvement in carcinoma treatment. Unfortunately, the scenario is different for sarcomas, a group of malignant neoplasms originating from mesenchymal cells, for which the main therapeutic approach still consists in the combination of surgery, chemotherapy, and radiation therapy. The lack of innovative approaches in sarcoma treatment stems from the high degree of heterogeneity of this tumor type, with more that 70 different histopathological subtypes, and the limited knowledge of the molecular drivers of tumor development and progression. Currently, molecular therapies are available mainly for the treatment of gastrointestinal stromal tumor, a soft-tissue malignancy characterized by an activating mutation of the tyrosine kinase KIT. Since the first application of this approach, a strong effort has been made to understand sarcoma molecular alterations that can be potential targets for therapy. The low incidence combined with the high level of histopathological heterogeneity makes the development of clinical trials for sarcomas very challenging. For this reason, preclinical studies are needed to better understand tumor biology with the aim to develop new targeted therapeutics. Currently, these studies are mainly based on in vitro testing, since cell lines, and in particular patient-derived models, represent a reliable and easy to handle tool for investigation. In the present review, we summarize the most important models currently available in the field, focusing in particular on the three-dimensional spheroid/organoid model. This innovative approach for studying tumor biology better represents tissue architecture and cell-cell as well as cell-microenvironment crosstalk, which are fundamental steps for tumor cell proliferation and survival.

Keywords: drug screening; in vitro organoid culture; patient-derived in vitro model; personalized medicine; preclinical model; sarcoma; sarcoma treatment.

Figures

Figure 1
Figure 1
Differentiation of normal mesenchymal stem cells (A) and altered differentiation (B). (B) The difference between the two hypotheses, whereby the initiating aberration occurs either at a later stage of differentiation (hypothesis 1) or hits the stem cell (hypothesis 2). Modified from the study by Teicher (24).
Figure 2
Figure 2
Different approaches for 3D cell culture model development. (A) Cellular spheroids: single cells from primary or stable cell lines aggregate together forming 3D structures. (B) Organotypic coculture: epithelial cells are cocultured with stroma cells embedded in a supporting matrix. (C) Organotypic slice culture: tissue slices obtained from the whole organ or from fragments of it are directly cultivated ex vivo. (D) Tissue organoids (PD3D™): primary cells isolated from fresh tissue without prior cell enrichment are grown as 3D multicellular structures [Modified from Silvestri et al. (64)].
Figure 3
Figure 3
Different methods for 3D spheroids development and growth. (A) Non-adhesive surfaces: culture plates with modified surfaces to reduce cell adhesion stimulate cell aggregation and formation of 3D structures. (B) Spinner flasks: stirred or rotating vessels are used to prevent cell adhesion to the surface of the plate allowing 3D spheroids formation. (C) Hanging drop: cells seeded in small drops of medium form cellular aggregates at the tip of the drop due to gravity forces. (D) Microcarrier beads: cells adhere to and proliferate on the surface of natural or synthetic solid beads forming 3D structures. (E) Hydrogel matrices: cells are seeded into matrices of natural or synthetic origin forming 3D structures by single cells aggregation or by monoclonal cell growth. [Modified from Silvestri et al. (64)].
Figure 4
Figure 4
Sarcoma spheroids growing in Matrigel-based three-dimensional cell culture.

References

    1. Mastrangelo G, Coindre J-M, Ducimetière F, Dei Tos AP, Fadda E, Blay J-Y, et al. Incidence of soft tissue sarcoma and beyond: a population-based prospective study in 3 European regions. Cancer (2012) 118:5339–48.10.1002/cncr.27555
    1. Ryan CW, Desai J. The past, present, and future of cytotoxic chemotherapy and pathway-directed targeted agents for soft tissue sarcoma. Am Soc Clin Oncol Educ Book (2013) 33:e386–93.10.1200/EdBook_AM.2013.33.e386
    1. Patel SR, Benjamin RS. Weichgewebe- und Knochensarkome und Knochenmetastasen. In: Suttorp N, Dietel M, editors. Harrison’s Principles of Internal Medicine (Harrisons Innere Medizin). Berlin: ABWWissenschaftsverlag GmbH, The McGraw-Hill Companies, Inc. (2013). p. 876–81.
    1. Burningham Z, Hashibe M, Spector L, Schiffman JD. The epidemiology of sarcoma. Clin Sarcoma Res (2012) 2:14.10.1186/2045-3329-2-14
    1. Stiller CA, Trama A, Serraino D, Rossi S, Navarro C, Chirlaque MD, et al. Descriptive epidemiology of sarcomas in Europe: report from the RARECARE project. Eur J Cancer (2013) 49:684–95.10.1016/j.ejca.2012.09.011
    1. American Cancer Society. Special Section: Rare Cancers in Adults. Atlanta: American Cancer Society; (2017).
    1. The ESMO/European Sarcoma Network Working Group. Soft tissue and visceral sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol (2014) 25:iii102–12.10.1093/annonc/mdu254
    1. The ESMO/European Sarcoma Network Working Group. Bone sarcomas: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol (2014) 25:iii113–23.10.1093/annonc/mdu256
    1. The ESMO/European Sarcoma Network Working Group. Gastrointestinal stromal tumours: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol (2014) 25:iii21–6.10.1093/annonc/mdu255
    1. Gatta G, van der Zwan JM, Casali PG, Siesling S, Dei Tos AP, Kunkler I, et al. Rare cancers are not so rare: the rare cancer burden in Europe. Eur J Cancer (2011) 47:2493–511.10.1016/j.ejca.2011.08.008
    1. Schütte J, Bauer S, Brodowicz T, Grünwald V, Hofer S, Hohenberger P, et al. Weichgewebssarkome des Erwachsenen. (2017). Available from:
    1. Fletcher CDM, Krishnan Unni K, Mertens F, editors. WHO Classification of Tumours, Pathology and Genetics of Tumours of Soft Tissue and Bone. Lyon: (2002). p. 1–415.
    1. Jo VY, Fletcher CDM. WHO classification of soft tissue tumours: an update based on the 2013 (4th) edition. Pathology (2014) 46:95–104.10.1097/PAT.0000000000000050
    1. Curtiss P, Strazzulla LC, Friedman-Kien AE. An update on Kaposi’s sarcoma: epidemiology, pathogenesis and treatment. Dermatol Ther (Heidelb) (2016) 6:465–70.10.1007/s13555-016-0152-3
    1. Helman LJ, Meltzer P. Mechanisms of sarcoma development. Nat Rev Cancer (2003) 3:685–94.10.1038/nrc1168
    1. Dancsok AR, Asleh-Aburaya K, Nielsen TO. Advances in sarcoma diagnostics and treatment. Oncotarget (2017) 8:7068–93.10.18632/oncotarget.12548
    1. Binh MBN, Sastre-Garau X, Guillou L, de Pinieux G, Terrier P, Lagacé R, et al. MDM2 and CDK4 immunostainings are useful adjuncts in diagnosing well-differentiated and dedifferentiated liposarcoma subtypes: a comparative analysis of 559 soft tissue neoplasms with genetic data. Am J Surg Pathol (2005) 29:1340–7.10.1097/01.pas.0000170343.09562.39
    1. Italiano A, Bianchini L, Keslair F, Bonnafous S, Cardot-Leccia N, Coindre J-M, et al. HMGA2 is the partner of MDM2 in well-differentiated and dedifferentiated liposarcomas whereas CDK4 belongs to a distinct inconsistent amplicon. Int J Cancer (2008) 122:2233–41.10.1002/ijc.23380
    1. Morrison BA. Soft tissue sarcomas of the extremities. Proc (Bayl Univ Med Cent) (2003) 16:285–90.
    1. Hirota S, Isozaki K, Moriyama Y, Hashimoto K, Nishida T, Ishiguro S, et al. Gain-of-function mutations of c-kit in human gastrointestinal stromal tumors. Science (1998) 279:577–80.10.1126/science.279.5350.577
    1. Corless CL, Barnett CM, Heinrich MC. Gastrointestinal stromal tumours: origin and molecular oncology. Nat Rev Cancer (2011) 11:865–78.10.1038/nrc3143
    1. Kurtovic-Kozaric A, Kugic A, Hasic A, Beslija S, Ceric T, Pasic A, et al. Long-term outcome of GIST patients treated with delayed imatinib therapy. Eur J Cancer (2017) 78:118–21.10.1016/j.ejca.2017.03.024
    1. Mohseny AB, Hogendoorn PCW. Concise review: mesenchymal tumors: when stem cells go mad. Stem Cells (2011) 29:397–403.10.1002/stem.596
    1. Teicher BA. Searching for molecular targets in sarcoma. Biochem Pharmacol (2012) 84:1–10.10.1016/j.bcp.2012.02.009
    1. Danielson LS, Menendez S, Attolini CS-O, Guijarro MV, Bisogna M, Wei J, et al. A differentiation-based microRNA signature identifies leiomyosarcoma as a mesenchymal stem cell-related malignancy. Am J Pathol (2010) 177:908–17.10.2353/ajpath.2010.091150
    1. Matushansky I, Hernando E, Socci ND, Matos T, Mills J, Edgar MA, et al. A developmental model of sarcomagenesis defines a differentiation-based classification for liposarcomas. Am J Pathol (2008) 172:1069–80.10.2353/ajpath.2008.070284
    1. Cleton-Jansen A-M, Anninga JK, Briaire-de Bruijn IH, Romeo S, Oosting J, Egeler RM, et al. Profiling of high-grade central osteosarcoma and its putative progenitor cells identifies tumourigenic pathways. Br J Cancer (2009) 101:2064–2064.10.1038/sj.bjc.6605482
    1. Nielsen TO, West RB, Linn SC, Alter O, Knowling MA, O’Connell JX, et al. Molecular characterisation of soft tissue tumours: a gene expression study. Lancet (2002) 359:1301–7.10.1016/S0140-6736(02)08270-3
    1. Boeuf S, Kunz P, Hennig T, Lehner B, Hogendoorn P, Bovée J, et al. A chondrogenic gene expression signature in mesenchymal stem cells is a classifier of conventional central chondrosarcoma. J Pathol (2008) 216:158–66.10.1002/path.2389
    1. Miura M, Miura Y, Padilla-Nash HM, Molinolo AA, Fu B, Patel V, et al. Accumulated chromosomal instability in murine bone marrow mesenchymal stem cells leads to malignant transformation. Stem Cells (2006) 24:1095–103.10.1634/stemcells.2005-0403
    1. Li H, Fan X, Kovi RC, Jo Y, Moquin B, Konz R, et al. Spontaneous expression of embryonic factors and p53 point mutations in aged mesenchymal stem cells: a model of age-related tumorigenesis in mice. Cancer Res (2007) 67:10889–98.10.1158/0008-5472.CAN-07-2665
    1. Hahn WC, Counter CM, Lundberg AS, Beijersbergen RL, Brooks MW, Weinberg RA. Creation of human tumour cells with defined genetic elements. Nature (1999) 400:464–8.10.1038/22780
    1. Rich JN, Guo C, McLendon RE, Bigner DD, Wang XF, Counter CM. A genetically tractable model of human glioma formation. Cancer Res (2001) 61:3556–60.
    1. Shima Y, Okamoto T, Aoyama T, Yasura K, Ishibe T, Nishijo K, et al. In vitro transformation of mesenchymal stem cells by oncogenic H-rasVal12. Biochem Biophys Res Commun (2007) 353:60–6.10.1016/j.bbrc.2006.11.137
    1. Li N, Yang R, Zhang W, Dorfman H, Rao P, Gorlick R. Genetically transforming human mesenchymal stem cells to sarcomas: changes in cellular phenotype and multilineage differentiation potential. Cancer (2009) 115:4795–806.10.1002/cncr.24519
    1. Mohseny AB, Szuhai K, Romeo S, Buddingh EP, Briaire-de Bruijn I, de Jong D, et al. Osteosarcoma originates from mesenchymal stem cells in consequence of aneuploidization and genomic loss of Cdkn2. J Pathol (2009) 219:294–305.10.1002/path.2603
    1. Adhikari AS, Agarwal N, Wood BM, Porretta C, Ruiz B, Pochampally RR, et al. CD117 and Stro-1 identify osteosarcoma tumor-initiating cells associated with metastasis and drug resistance. Cancer Res (2010) 70:4602–12.10.1158/0008-5472.CAN-09-3463
    1. Levings PP, McGarry SV, Currie TP, Nickerson DM, McClellan S, Ghivizzani SC, et al. Expression of an exogenous human Oct-4 promoter identifies tumor-initiating cells in osteosarcoma. Cancer Res (2009) 69:5648–55.10.1158/0008-5472.CAN-08-3580
    1. Fujii H, Honoki K, Tsujiuchi T, Kido A, Yoshitani K, Takakura Y. Sphere-forming stem-like cell populations with drug resistance in human sarcoma cell lines. Int J Oncol (2009) 34:1381–6.10.3892/ijo_00000265
    1. Honoki K. Do stem-like cells play a role in drug resistance of sarcomas? Expert Rev Anticancer Ther (2010) 10:261–70.10.1586/era.09.184
    1. Italiano A, Di Mauro I, Rapp J, Pierron G, Auger N, Alberti L, et al. Clinical effect of molecular methods in sarcoma diagnosis (GENSARC): a prospective, multicentre, observational study. Lancet Oncol (2016) 17:532–8.10.1016/S1470-2045(15)00583-5
    1. Gerlinger M, Horswell S, Larkin J, Rowan AJ, Salm MP, Varela I, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet (2014) 46:225–33.10.1038/ng.2891
    1. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med (2012) 366:883–92.10.1056/NEJMoa1113205
    1. Rye IH, Helland Å, Saetersdal A, Naume B, Almendro V, Polyak K, et al. Intra-tumor heterogeneity as a predictor of therapy response in HER2 positive breast cancer. Cancer Res (2012) 7210.1158/0008-5472.SABCS12-P3-05-04
    1. Tougeron D, Lecomte T, Pages JC, Villalva C, Collin C, Ferru A, et al. Effect of low-frequency KRAS mutations on the response to anti-EGFR therapy in metastatic colorectal cancer. Ann Oncol (2013) 24:1267–73.10.1093/annonc/mds620
    1. Bai H, Wang Z, Wang Y, Zhuo M, Zhou Q, Duan J, et al. Detection and clinical significance of intratumoral EGFR mutational heterogeneity in Chinese patients with advanced non-small cell lung cancer. PLoS One (2013) 8:e54170.10.1371/journal.pone.0054170
    1. Patwardhan PP, Surriga O, Beckman MJ, de Stanchina E, Dematteo RP, Tap WD, et al. Sustained inhibition of receptor tyrosine kinases and macrophage depletion by PLX3397 and rapamycin as a potential new approach for the treatment of MPNSTs. Clin Cancer Res (2014) 20:3146–58.10.1158/1078-0432.CCR-13-2576
    1. Salawu A, Fernando M, Hughes D, Reed MWR, Woll P, Greaves C, et al. Establishment and molecular characterisation of seven novel soft-tissue sarcoma cell lines. Br J Cancer (2016) 115:1058–68.10.1038/bjc.2016.259
    1. Bruland O, Fodstad O, Pihl A. The use of multicellular spheroids in establishing human sarcoma cell lines in vitro. Int J Cancer (1985) 35:793–8.10.1002/ijc.2910350616
    1. Hu M, Nicolson GL, Trent JC, Yu D, Zhang L, Lang A, et al. Characterization of 11 human sarcoma cell strains. Cancer (2002) 95:1569–76.10.1002/cncr.10879
    1. Ottaviano L, Schaefer K-L, Gajewski M, Huckenbeck W, Baldus S, Rogel U, et al. Molecular characterization of commonly used cell lines for bone tumor research: a trans-European EuroBoNet effort. Genes Chromosomes Cancer (2010) 49:40–51.10.1002/gcc.20717
    1. Mohseny AB, Machado I, Cai Y, Schaefer K-L, Serra M, Hogendoorn PCW, et al. Functional characterization of osteosarcoma cell lines provides representative models to study the human disease. Lab Invest (2011) 91:1195–205.10.1038/labinvest.2011.72
    1. Kenny PA, Lee GY, Myers CA, Neve RM, Semeiks JR, Spellman PT, et al. The morphologies of breast cancer cell lines in three-dimensional assays correlate with their profiles of gene expression. Mol Oncol (2007) 1:84–96.10.1016/j.molonc.2007.02.004
    1. Silberstein GB. Tumour-stromal interactions. Role of the stroma in mammary development. Breast Cancer Res (2001) 3:218–23.10.1186/bcr299
    1. Schmeichel KL, Bissell MJ. Modeling tissue-specific signaling and organ function in three dimensions. J Cell Sci (2003) 116:2377–88.10.1242/jcs.00503
    1. Akkerman N, Defize LHK. Dawn of the organoid era: 3D tissue and organ cultures revolutionize the study of development, disease, and regeneration. Bioessays (2017) 39:1600244.10.1002/bies.201600244
    1. Bai C, Yang M, Fan Z, Li S, Gao T, Fang Z. Associations of chemo- and radio-resistant phenotypes with the gap junction, adhesion and extracellular matrix in a three-dimensional culture model of soft sarcoma. J Exp Clin Cancer Res (2015) 34:58.10.1186/s13046-015-0175-0
    1. Salerno M, Avnet S, Bonuccelli G, Eramo A, De Maria R, Gambarotti M, et al. Sphere-forming cell subsets with cancer stem cell properties in human musculoskeletal sarcomas. Int J Oncol (2013) 43:95–102.10.3892/ijo.2013.1927
    1. Fong ELS, Lamhamedi-Cherradi S-E, Burdett E, Ramamoorthy V, Lazar AJ, Kasper FK, et al. Modeling Ewing sarcoma tumors in vitro with 3D scaffolds. Proc Natl Acad Sci U S A (2013) 110:6500–5.10.1073/pnas.1221403110
    1. Villasante A, Marturano-Kruik A, Robinson ST, Liu Z, Guo XE, Vunjak-Novakovic G. Tissue-engineered model of human osteolytic bone tumor. Tissue Eng Part C Methods (2017) 23:98–107.10.1089/ten.TEC.2016.0371
    1. Yuhas JM, Li AP, Martinez AO, Ladman AJ. A simplified method for production and growth of multicellular tumor spheroids. Cancer Res (1977) 37:3639–43.
    1. Bae SI, Kang GH, Kim YI, Lee BL, Kleinman HK, Kim WH. Development of intracytoplasmic lumens in a colon cancer cell line cultured on a non-adhesive surface. Cancer Biochem Biophys (1999) 17:35–47.10.1016/S0940-2993(99)80053-0
    1. Lin R-Z, Lin R-Z, Chang H-Y. Recent advances in three-dimensional multicellular spheroid culture for biomedical research. Biotechnol J (2008) 3:1172–84.10.1002/biot.200700228
    1. Silvestri A, Schumacher D, Silvestrov M, Schäfer R, Reinhard C, Hoffmann J, et al. In vitro three-dimensional cell cultures as tool for precision medicine. In: Haybaeck J, editor. Mechanisms of Molecular Carcinogenesis. (Vol. 2), Cham: Springer International Publishing; (2017). p. 281–313.
    1. Sutherland RM, Inch WR, McCredie JA, Kruuv J. A multi-component radiation survival curve using an in vitro tumour model. Int J Radiat Biol Relat Stud Phys Chem Med (1970) 18:491–5.10.1080/09553007014551401
    1. Kelm JM, Timmins NE, Brown CJ, Fussenegger M, Nielsen LK. Method for generation of homogeneous multicellular tumor spheroids applicable to a wide variety of cell types. Biotechnol Bioeng (2003) 83:173–80.10.1002/bit.10655
    1. Johns RA, Tichotsky A, Muro M, Spaeth JP, Le Cras TD, Rengasamy A. Halothane and isoflurane inhibit endothelium-derived relaxing factor-dependent cyclic guanosine monophosphate accumulation in endothelial cell-vascular smooth muscle co-cultures independent of an effect on guanylyl cyclase activation. Anesthesiology (1995) 83:823–34.10.1097/00000542-199510000-00023
    1. Bell E. Strategy for the selection of scaffolds for tissue engineering. Tissue Eng (1995) 1:163–79.10.1089/ten.1995.1.163
    1. Hughes CS, Postovit LM, Lajoie GA. Matrigel: a complex protein mixture required for optimal growth of cell culture. Proteomics (2010) 10:1886–90.10.1002/pmic.200900758
    1. Abu-Yousif AO, Rizvi I, Evans CL, Celli JP, Hasan T. PuraMatrix encapsulation of cancer cells. J Vis Exp (2009) (34):e1692.10.3791/1692
    1. Mackenzie IC, Fusenig NE. Regeneration of organized epithelial structure. J Invest Dermatol (1983) 81:S189–94.10.1111/1523-1747.ep12541093
    1. Stark HJ, Baur M, Breitkreutz D, Mirancea N, Fusenig NE. Organotypic keratinocyte cocultures in defined medium with regular epidermal morphogenesis and differentiation. J Invest Dermatol (1999) 112:681–91.10.1046/j.1523-1747.1999.00573.x
    1. Barton CE, Johnson KN, Mays DM, Boehnke K, Shyr Y, Boukamp P, et al. Novel p63 target genes involved in paracrine signaling and keratinocyte differentiation. Cell Death Dis (2010) 1:e74.10.1038/cddis.2010.49
    1. Boehnke K, Mirancea N, Pavesio A, Fusenig NE, Boukamp P, Stark H-J. Effects of fibroblasts and microenvironment on epidermal regeneration and tissue function in long-term skin equivalents. Eur J Cell Biol (2007) 86:731–46.10.1016/j.ejcb.2006.12.005
    1. van der Kuip H, Mürdter TE, Sonnenberg M, McClellan M, Gutzeit S, Gerteis A, et al. Short term culture of breast cancer tissues to study the activity of the anticancer drug taxol in an intact tumor environment. BMC Cancer (2006) 6:86.10.1186/1471-2407-6-86
    1. Estes JM, Oliver PG, Straughn JM, Zhou T, Wang W, Grizzle WE, et al. Efficacy of anti-death receptor 5 (DR5) antibody (TRA-8) against primary human ovarian carcinoma using a novel ex vivo tissue slice model. Gynecol Oncol (2007) 105:291–8.10.1016/j.ygyno.2006.12.033
    1. Kiviharju-af Hällström TM, Jäämaa S, Mönkkönen M, Peltonen K, Andersson LC, Medema RH, et al. Human prostate epithelium lacks Wee1A-mediated DNA damage-induced checkpoint enforcement. Proc Natl Acad Sci U S A (2007) 104:7211–6.10.1073/pnas.0609299104
    1. Kondo J, Endo H, Okuyama H, Ishikawa O, Iishi H, Tsujii M, et al. Retaining cell-cell contact enables preparation and culture of spheroids composed of pure primary cancer cells from colorectal cancer. Proc Natl Acad Sci U S A (2011) 108:6235–40.10.1073/pnas.1015938108
    1. Endo H, Okami J, Okuyama H, Kumagai T, Uchida J, Kondo J, et al. Spheroid culture of primary lung cancer cells with neuregulin 1/HER3 pathway activation. J Thorac Oncol (2013) 8:131–9.10.1097/JTO.0b013e3182779ccf
    1. Ashley N, Jones M, Ouaret D, Wilding J, Bodmer WF. Rapidly derived colorectal cancer cultures recapitulate parental cancer characteristics and enable personalized therapeutic assays. J Pathol (2014) 234:34–45.10.1002/path.4371
    1. Sachs N, Clevers H. Organoid cultures for the analysis of cancer phenotypes. Curr Opin Genet Dev (2014) 24:68–73.10.1016/j.gde.2013.11.012
    1. Boehnke K, Iversen PW, Schumacher D, Lallena MJ, Haro R, Amat J, et al. Assay establishment and validation of a high-throughput screening platform for three-dimensional patient-derived colon cancer organoid cultures. J Biomol Screen (2016) 21:931–41.10.1177/1087057116650965
    1. Schütte M, Risch T, Abdavi-Azar N, Boehnke K, Schumacher D, Keil M, et al. Molecular dissection of colorectal cancer in pre-clinical models identifies biomarkers predicting sensitivity to EGFR inhibitors. Nat Commun (2017) 8:14262.10.1038/ncomms14262
    1. Barretina J, Taylor BS, Banerji S, Ramos AH, Lagos-Quintana M, DeCarolis PL, et al. Subtype-specific genomic alterations define new targets for soft-tissue sarcoma therapy. Nat Genet (2010) 42:715–21.10.1038/ng.619
    1. Jour G, Scarborough JD, Jones RL, Loggers E, Pollack SM, Pritchard CC, et al. Molecular profiling of soft tissue sarcomas using next-generation sequencing: a pilot study toward precision therapeutics. Hum Pathol (2014) 45:1563–71.10.1016/j.humpath.2014.04.012
    1. Hanes R, Grad I, Lorenz S, Stratford EW, Munthe E, Reddy CCS, et al. Preclinical evaluation of potential therapeutic targets in dedifferentiated liposarcoma. Oncotarget (2016) 7:54583–95.10.18632/oncotarget.10518
    1. Treindl F, Ruprecht B, Beiter Y, Schultz S, Döttinger A, Staebler A, et al. A bead-based western for high-throughput cellular signal transduction analyses. Nat Commun (2016) 7:12852.10.1038/ncomms12852
    1. Suehara Y, Kondo T, Fujii K, Hasegawa T, Kawai A, Seki K, et al. Proteomic signatures corresponding to histological classification and grading of soft-tissue sarcomas. Proteomics (2006) 6:4402–9.10.1002/pmic.200600196
    1. Suehara Y, Kondo T, Seki K, Shibata T, Fujii K, Gotoh M, et al. Pfetin as a prognostic biomarker of gastrointestinal stromal tumors revealed by proteomics. Clin Cancer Res (2008) 14:1707–17.10.1158/1078-0432.CCR-07-1478
    1. Petricoin EF, Espina V, Araujo RP, Midura B, Yeung C, Wan X, et al. Phosphoprotein pathway mapping: Akt/mammalian target of rapamycin activation is negatively associated with childhood rhabdomyosarcoma survival. Cancer Res (2007) 67:3431–40.10.1158/0008-5472.CAN-06-1344
    1. Kondo T, Kawai A. A proteomics approach for the development of sarcoma biomarkers. EuPA Open Proteom (2014) 4:121–8.10.1016/j.euprot.2014.06.004
    1. Grohar PJ, Woldemichael GM, Griffin LB, Mendoza A, Chen Q-R, Yeung C, et al. Identification of an inhibitor of the EWS-FLI1 oncogenic transcription factor by high-throughput screening. J Natl Cancer Inst (2011) 103:962–78.10.1093/jnci/djr156
    1. Boro A, Prêtre K, Rechfeld F, Thalhammer V, Oesch S, Wachtel M, et al. Small-molecule screen identifies modulators of EWS/FLI1 target gene expression and cell survival in Ewing’s sarcoma. Int J Cancer (2012) 131:2153–64.10.1002/ijc.27472
    1. Teicher BA, Polley E, Kunkel M, Evans D, Silvers T, Delosh R, et al. Sarcoma cell line screen of oncology drugs and investigational agents identifies patterns associated with gene and microRNA expression. Mol Cancer Ther (2015) 14:2452–62.10.1158/1535-7163.MCT-15-0074
    1. Apple S. Starving the Beast. New York: The New York Times; (2016). MM64 p.
    1. Schütte M, Ogilvie LA, Rieke DT, Lange BMH, Yaspo M-L, Lehrach H. Cancer precision medicine: why more is more and DNA is not enough. Public Health Genomics (2017).10.1159/000477157
    1. Pauli C, Hopkins BD, Prandi D, Shaw R, Fedrizzi T, Sboner A, et al. Personalized in vitro and in vivo cancer models to guide precision medicine. Cancer Discov (2017) 7:462–77.10.1158/-16-1154
    1. Chakradhar S. Put to the test: organoid-based testing becomes a clinical tool. Nat Med (2017) 23:796–9.10.1038/nm0717-796

Source: PubMed

3
Subscribe