Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Application of Three-dimensional (3D) Tumor Cell Culture Systems and Mechanism of Drug Resistance

Author(s): Adeeb Shehzad, Vijaya Ravinayagam, Hamad AlRumaih, Meneerah Aljafary, Dana Almohazey, Sarah Almofty, Noor A. Al-Rashid and Ebtesam A. Al-Suhaimi*

Volume 25, Issue 34, 2019

Page: [3599 - 3607] Pages: 9

DOI: 10.2174/1381612825666191014163923

Price: $65

Abstract

The in-vitro experimental model for the development of cancer therapeutics has always been challenging. Recently, the scientific revolution has improved cell culturing techniques by applying three dimensional (3D) culture system, which provides a similar physiologically relevant in-vivo model for studying various diseases including cancer. In particular, cancer cells exhibiting in-vivo behavior in a model of 3D cell culture is a more accurate cell culture model to test the effectiveness of anticancer drugs or characterization of cancer cells in comparison with two dimensional (2D) monolayer. This study underpins various factors that cause resistance to anticancer drugs in forms of spheroids in 3D in-vitro cell culture and also outlines key challenges and possible solutions for the future development of these systems.

Keywords: Spheroid, 3D cell culture, drug resistance, drug discovery, cancer therapy, 2D monolayer.

[1]
Zips D, Thames HD, Baumann M. New anticancer agents: in vitro and in vivo evaluation. In Vivo 2005; 19: 1-7.
[2]
Jaroch K, Jaroch A, Bojko B. Cell cultures in drug discovery and development: the need of reliable in vitro-in vivo extrapolation for pharmacodynamics and pharmacokinetics assessment. J Pharm Biomed Anal 2018; 147: 297-312.
[http://dx.doi.org/10.1016/j.jpba.2017.07.023]
[3]
Langhans SA. Three-dimensional in vitro cell culture models in drug discovery and drug repositioning. Front Pharmacol 2018; 9: 6.
[http://dx.doi.org/10.3389/fphar.2018.00006]
[4]
Aggarwal BB, Danda D, Gupta S, Gehlot P. Models for prevention and treatment of cancer: problems vs. promises. Biochem Pharmacol 2009; 78: 1083-94.
[http://dx.doi.org/10.1016/j.bcp.2009.05.027]
[5]
Aljitawi OS, Li D, Xiao Y, et al. A novel three-dimensional stromal-based model for in vitro chemotherapy sensitivity testing of leukemia cells. Leuk Lymphoma 2014; 55: 378-91.
[http://dx.doi.org/10.3109/10428194.2013.793323]
[6]
Antoni D, Burckel H. Josset, Noel G. Three-dimensional cell culture: a breakthrough in-vivo. Int J Mol Sci 2015; 16: 5517-27.
[http://dx.doi.org/10.3390/ijms16035517]
[7]
Verjans ET, Doijen J, Luyten W, Landuyt B, Schoofs L. Three-dimensional cell culture models for anticancer drug screening: worth the effort? J Cell Physiol 2018; 233: 2993-3003.
[http://dx.doi.org/10.1002/jcp.26052]
[8]
Barbone D, Ryan J, Kolhatkar N, et al. The Bcl-2 repertoire of mesothelioma spheroids underlies acquired apoptotic multicellular resistance. Cell Death Dis 2011; 2e174
[http://dx.doi.org/10.1038/cddis.2011.58]
[9]
Singhera F, Cooper E, Scampavia L, Spicer T. Using bead injection to model dispensing of 3-D multicellular spheroids into microtiter plates. Talanta 2018; 177: 74-6.
[http://dx.doi.org/10.1016/j.talanta.2017.09.022]
[10]
Fang Y, Eglen RM. Three-dimensional cell cultures in drug discovery and development. SLAS Discov 2017; 22: 456-72.
[http://dx.doi.org/10.1177/1087057117696795]
[11]
Chambers KF, Mosaad EM, Russell PJ, Clements JA, Doran MR. 3D cultures of prostate cancer cells cultured in a novel high-throughput culture platform are more resistant to chemotherapeutics compared to cells cultured in monolayer. PLoS One 2014; 9e111029
[http://dx.doi.org/10.1371/journal.pone.0111029]
[12]
Ishiguro T, Ohata H, Sato A, Yamawaki K, Enomoto T, Okamoto K. Tumor‐derived spheroids: relevance to cancer stem cells and clinical applications. Cancer Sci 2017; 108: 283-9.
[http://dx.doi.org/10.1111/cas.13155]
[13]
Schubert C, Van Langeveld MC, Donoso LA. Innovations in 3D printing: a 3D overview from optics to organs. Br J Ophthalmol 2014; 98: 159-61.
[http://dx.doi.org/10.1136/bjophthalmol-2013-304446]
[14]
Shehzad A, Wahid F, Lee YS. Curcumin in cancer chemoprevention: molecular targets, pharmacokinetics, bioavailability, and clinical trials. Arch Pharm (Weinheim) 2010; 343: 489-99.
[http://dx.doi.org/10.1002/ardp.200900319]
[15]
Decaup E, Jean C, Laurent C, et al. Anti-tumor activity of obinutuzumab and rituximab in a follicular lymphoma 3D model. Blood Cancer J 2013; 3e131
[http://dx.doi.org/10.1038/bcj.2013.32]
[16]
Fromigue O, Louis K, Dayem M, et al. Gene expression profiling of normal human pulmonary fibroblasts following coculture with non-small-cell lung cancer cells reveals alterations related to matrix degradation, angiogenesis, cell growth and survival. Oncogene 2003; 22: 8487-97.
[http://dx.doi.org/10.1038/sj.onc.1206918]
[17]
Friedland JC, Lakins JN, Kazanietz MG, Chernoff J, Boettiger D, Weaver VM. α6β4 integrin activates Rac-dependent p21-activated kinase 1 to drive NF-κB-dependent resistance to apoptosis in 3D mammary acini. J Cell Sci 2007; 120: 3700-12.
[http://dx.doi.org/10.1242/jcs.03484]
[18]
Lee SK, Shehzad A, Jung JC, et al. Protein kinase Cα protects against multidrug resistance in human colon cancer cells. Mol Cells 2012; 34: 61-9.
[http://dx.doi.org/10.1007/s10059-012-0087-1]
[19]
Shehzad A, Rehman G, Ul-Islam M, Khattak WA, Lee YS. Challenges in the development of drugs for the treatment of tuberculosis. Braz J Infect Dis 2013; 17: 74-81.
[http://dx.doi.org/10.1016/j.bjid.2012.10.009]
[20]
Shehzad A, Lee J, Lee YS. Curcumin in various cancers. Biofactors 2013; 39: 56-68.
[http://dx.doi.org/10.1002/biof.1068]
[21]
Wahid F, Khan T, Shehzad A, Ui-Islam M, Kim YY. Interaction of nanomaterials with cells and their medical applications. J Nanosci Nanotechnol 2014; 14: 744-54.
[http://dx.doi.org/10.1166/jnn.2014.9016]
[22]
Hsieh CH, Chen YD, Huang SF, Wang HM, Wu MH. The effect of primary cancer cell culture models on the results of drug chemosensitivity assays: the application of perfusion microbioreactor system as cell culture vessel. BioMed Res Int 2015; 2015470283
[http://dx.doi.org/10.1155/2015/470283]
[23]
Edmondson R, Broglie JJ, Adcock AF, Yang L. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol 2014; 12: 207-18.
[http://dx.doi.org/10.1089/adt.2014.573]
[24]
Hoarau-Véchot J, Rafii A, Touboul C, Pasquier J. Halfway between 2D and animal models: are 3D cultures the ideal tool to study cancer-microenvironment interactions? Int J Mol Sci 2018; 19: 181.
[http://dx.doi.org/10.3390/ijms19010181]
[25]
Gaebler M, Silvestri A, Haybaeck J, et al. Three-dimensional patient- derived in vitro sarcoma models: promising tools for improving clinical tumor management. Front Oncol 2017; 11; 7: 203.
[26]
Ito A, Ino K, Kobayashi T, Honda H. The effect of RGD peptide-conjugated magnetite cationic liposomes on cell growth and cell sheet harvesting. Biomaterials 2005; 26: 6185-93.
[http://dx.doi.org/10.1016/j.biomaterials.2005.03.039]
[27]
Ku SH, Ryu J, Hong SK, Lee H, Park CB. General functionalization route for cell adhesion on non-wetting surfaces. Biomaterials 2010; 31: 2535-41.
[http://dx.doi.org/10.1016/j.biomaterials.2009.12.020]
[28]
Pampaloni F, Stelzer EH. Three-dimensional cell cultures in toxicology. Biotechnol Genet Eng Rev 2009; 26: 117-38.
[http://dx.doi.org/10.5661/bger-26-117]
[29]
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.
[http://dx.doi.org/10.1002/bit.10655]
[30]
Costa EC, Moreira AF, de Melo-Diogo D, Gaspar VM, Carvalho MP, Correia IJ. 3D tumor spheroids: an overview on the tools and techniques used for their analysis. Biotechnol Adv 2016; 34: 1427-41.
[http://dx.doi.org/10.1016/j.biotechadv.2016.11.002]
[31]
Bokhari MA, Akay G, Zhang S, Birch MA. The enhancement of osteoblast growth and differentiation in vitro on a peptide hydrogel-polyHIPE polymer hybrid material. Biomaterials 2005; 26: 5198-208.
[http://dx.doi.org/10.1016/j.biomaterials.2005.01.040]
[32]
Lee BH, Kim MH, Lee JH, Seliktar D, Cho NJ, Tan LP. Modulation of Huh7.5 spheroid formation and functionality using modified PEG-based hydrogels of different stiffness. PLoS One 2015; 10e0118123
[http://dx.doi.org/10.1371/journal.pone.0118123]
[33]
Shehzad A, Ul-Islam M, Wahid F, Lee YS. Multifunctional polymeric nanocurcumin for cancer therapy. J Nanosci Nanotechnol 2014; 14: 803-14.
[http://dx.doi.org/10.1166/jnn.2014.9103]
[34]
Nam KH, Smith AS, Lone S, Kwon S, Kim DH. Biomimetic 3D tissue models for advanced high-throughput drug screening. J Lab Autom 2015; 20: 201-15.
[http://dx.doi.org/10.1177/2211068214557813]
[35]
Yoshii Y, Furukawa T, Waki A, et al. High-throughput screening with nanoimprinting 3D culture for efficient drug development by mimicking the tumor environment. Biomaterials 2015; 51: 278-89.
[http://dx.doi.org/10.1016/j.biomaterials.2015.02.008]
[36]
Martinez NJ, Titus SA, Wagner AK, Simeonov A. High-throughput fluorescence imaging approaches for drug discovery using in vitro and in vivo three-dimensional models. Expert Opin Drug Discov 2015; 10: 1347-61.
[http://dx.doi.org/10.1517/17460441.2015.1091814]
[37]
Yoshii Y, Furukawa T, Aoyama H, et al. Regorafenib as a potential adjuvant chemotherapy agent in disseminated small colon cancer: drug selection outcome of a novel screening system using nanoimprinting 3-dimensional culture with HCT116-RFP cells. Int J Oncol 2016; 48: 1477-84.
[http://dx.doi.org/10.3892/ijo.2016.3361]
[38]
Wang JZ, Zhu YX, Ma HC, et al. Developing multi-cellular tumor spheroid model (MCTS) in the chitosan/collagen/alginate (CCA) fibrous scaffold for anticancer drug screening. Mater Sci Eng C 2016; 62: 215-25.
[http://dx.doi.org/10.1016/j.msec.2016.01.045]
[39]
Cavo M, Fato M, Penuela L, Beltrame F, Raiteri R, Scaglione S. Microenvironment complexity and matrix stiffness regulate breast cancer cell activity in a 3D in vitro model. Sci Rep 2016; 6: 35367.
[http://dx.doi.org/10.1038/srep35367]
[40]
Chen L, Xiao Z, Meng Y, et al. The enhancement of cancer stem cell properties of MCF-7 cells in 3D collagen scaffolds for modeling of cancer and anti-cancer drugs. Biomaterials 2012; 33: 1437-44.
[http://dx.doi.org/10.1016/j.biomaterials.2011.10.056]
[41]
Al-Suhaimi EA1. Shehzad A. Leptin, resistin and visfatin: the missing link between endocrine metabolic disorders and immunity. Eur J Med Res 2013; 18: 12.
[http://dx.doi.org/10.1186/2047-783X-18-12]
[42]
Eder T, Weber A, Neuwirt H, et al. Cancer-associated fibroblasts modify the response of prostate cancer cells to androgen and anti-androgens in three-dimensional spheroid culture. Int J Mol Sci 2016; 17: 1458.
[http://dx.doi.org/10.3390/ijms17091458]
[43]
Jaganathan H, Gage J, Leonard F, et al. Three-dimensional in vitro co-culture model of breast tumor using magnetic levitation. Sci Rep 2014; 4: 6468.
[http://dx.doi.org/10.1038/srep06468]
[44]
Brancato V, Comunanza V, Imparato G, et al. Bioengineered tumoral microtissues recapitulate desmoplastic reaction of pancreatic cancer. Acta Biomater 2017; 49: 152-66.
[http://dx.doi.org/10.1016/j.actbio.2016.11.072]
[45]
Santo VE, Estrada MF, Rebelo SP, et al. Adaptable stirred-tank culture strategies for large scale production of multicellular spheroid-based tumor cell models. J Biotechnol 2016; 221: 118-29.
[http://dx.doi.org/10.1016/j.jbiotec.2016.01.031]
[46]
Chung M, Ahn J, Son K, Kim S, Jeon NL. Biomimetic model of tumor microenvironment on microfluidic platform. Adv Healthc Mater 2017; •••: 6.
[http://dx.doi.org/10.1002/adhm.201700196]
[47]
Hsiao AY, Torisawa YS, Tung YC, et al. Microfluidic system for formation of PC-3 prostate cancer co-culture spheroids. Biomaterials 2009; 30: 3020-7.
[http://dx.doi.org/10.1016/j.biomaterials.2009.02.047]
[48]
Zhang Z, Nagrath S. Microfluidics and cancer: are we there yet? Biomed Microdevices 2013; 15: 595-609.
[http://dx.doi.org/10.1007/s10544-012-9734-8]
[49]
Islam SU, Ahmed MB, Lee SJ, et al. PRP4 kinase induces actin rearrangement and epithelial-mesenchymal transition through modulation of the actin-binding protein cofilin. Exp Cell Res 2018; 369: 158-65.
[http://dx.doi.org/10.1016/j.yexcr.2018.05.018]
[50]
Shehzad A, Qureshi M, Anwar MN, Lee YS. Multifunctional curcumin mediate multitherapeutic effects. J Food Sci 2017; 82: 2006-15.
[http://dx.doi.org/10.1111/1750-3841.13793]
[51]
Shehzad A, Ul Islam S, Lee J, Lee YS. Prostaglandin E2 reverses curcumin-induced inhibition of survival signal pathways in human colorectal carcinoma (HCT-15) cell lines. Mol Cells 2014; 37: 899-906.
[http://dx.doi.org/10.14348/molcells.2014.0212]
[52]
Shehzad A, Park JW, Lee J, Lee YS. Curcumin induces radiosensitivity of in vitro and in vivo cancer models by modulating pre-mRNA processing factor 4 (Prp4). Chem Biol Interact 2013; 206: 394-402.
[http://dx.doi.org/10.1016/j.cbi.2013.10.007]
[53]
Loessner D, Stok KS, Lutolf MP, Hutmacher DW, Clements JA, Rizzi SC. Bioengineered 3D platform to explore cell-ECM interactions and drug resistance of epithelial ovarian cancer cells. Biomaterials 2010; 31: 8494-506.
[http://dx.doi.org/10.1016/j.biomaterials.2010.07.064]
[54]
Chen YC, Zhang Z, Fouladdel S, et al. Single cell dual adherent-suspension co-culture micro-environment for studying tumor-stromal interactions with functionally selected cancer stem-like cells. Lab Chip 2016; 16: 2935-45.
[http://dx.doi.org/10.1039/C6LC00062B]
[55]
Rafehi S, Ramos Valdes Y, Bertrand M, et al. TGFβ signaling regulates epithelial-mesenchymal plasticity in ovarian cancer ascites-derived spheroids. Endocr Relat Cancer 2016; 23: 147-59.
[http://dx.doi.org/10.1530/ERC-15-0383]
[56]
Desjardins M, Xie J, Gurler H, et al. Versican regulates metastasis of epithelial ovarian carcinoma cells and spheroids. J Ovarian Res 2014; 7: 70.
[http://dx.doi.org/10.1186/1757-2215-7-70]
[57]
Al Habyan S, Kalos C, Szymborski J, McCaffrey L. Multicellular detachment generates metastatic spheroids during intra-abdominal dissemination in epithelial ovarian cancer. Oncogene 2018; 37: 5127-35.
[http://dx.doi.org/10.1038/s41388-018-0317-x]
[58]
Bersini S, Jeon JS, Dubini G, et al. A microfluidic 3D in vitro model for specificity of breast cancer metastasis to bone. Biomaterials 2014; 35: 2454-61.
[http://dx.doi.org/10.1016/j.biomaterials.2013.11.050]
[59]
Wang X, Sun L, Maffini MV, Soto A, Sonnenschein C, Kaplan DL. A complex 3D human tissue culture system based on mammary stromal cells and silk scaffolds for modeling breast morphogenesis and function. Biomaterials 2010; 31: 3920-9.
[http://dx.doi.org/10.1016/j.biomaterials.2010.01.118]
[60]
Hongisto V, Jernström S, Fey V, et al. High-throughput 3D screening reveals differences in drug sensitivities between culture models of JIMT1 breast cancer cells. PLoS One 2013; 8e77232
[http://dx.doi.org/10.1371/journal.pone.0077232]
[61]
Hirschhaeuser F, Menne H, Dittfeld C, West J, Mueller-Klieser W, Kunz-Schughart LA. Multicellular tumor spheroids: an underestimated tool is catching up again. J Biotechnol 2010; 148: 3-15.
[http://dx.doi.org/10.1016/j.jbiotec.2010.01.012]
[62]
Liang D, Ma Y, Liu J, et al. The hypoxic microenvironment upgrades stem-like properties of ovarian cancer cells. BMC Cancer 2012; 12: 201.
[http://dx.doi.org/10.1186/1471-2407-12-201]
[63]
Lovitt CJ, Shelper TB, Avery VM. Advanced cell culture techniques for cancer drug discovery. Biology (Basel) 2014; 3: 345-67.
[http://dx.doi.org/10.3390/biology3020345]
[64]
Lee JM, Mhawech-Fauceglia P, Lee N, et al. A three-dimensional microenvironment alters protein expression and chemosensitivity of epithelial ovarian cancer cells in vitro. Lab Invest 2013; 93: 528.
[http://dx.doi.org/10.1038/labinvest.2013.41]
[65]
Liu C, Liu Y, Xu XX, et al. Potential effect of matrix stiffness on the enrichment of tumor initiating cells under three-dimensional culture conditions. Exp Cell Res 2015; 330: 123-34.
[http://dx.doi.org/10.1016/j.yexcr.2014.07.036]
[66]
Liu J, Ma L, Xu J, et al. Spheroid body-forming cells in the human gastric cancer cell line MKN-45 possess cancer stem cell properties. Int J Oncol 2013; 42: 453-9.
[http://dx.doi.org/10.3892/ijo.2012.1720]
[67]
Shehzad A, Parveen S, Qureshi M, Subhan F, Lee YS. Decursin and decursinol angelate: molecular mechanism and therapeutic potential in inflammatory diseases. Inflamm Res 2018; 67: 209-18.
[http://dx.doi.org/10.1007/s00011-017-1114-7]
[68]
Leng Z, Tao K, Xia Q, et al. Krüppel-like factor 4 acts as an oncogene in colon cancer stem cell-enriched spheroid cells. PLoS One 2013; 8e56082
[http://dx.doi.org/10.1371/journal.pone.0056082]
[69]
Qureshi M, Al-Suhaimi EA, Wahid F, Shehzad O, Shehzad A. Therapeutic potential of curcumin for multiple sclerosis. Neurol Sci 2018; 39: 207-14.
[http://dx.doi.org/10.1007/s10072-017-3149-5]
[70]
Mehta G, Hsiao AY, Ingram M, Luker GD, Takayama S. Opportunities and challenges for use of tumor spheroids as models to test drug delivery and efficacy. J Control Release 2012; 164: 192-204.
[http://dx.doi.org/10.1016/j.jconrel.2012.04.045]
[71]
Nyga A, Cheema U, Loizidou M. 3D tumour models: novel in vitro approaches to cancer studies. J Cell Commun Signal 2011; 5: 239-48.
[http://dx.doi.org/10.1007/s12079-011-0132-4]
[72]
Petersen MC, Lazar J, Jacob HJ, Wakatsuki T. Tissue engineering: a new frontier in physiological genomics. Physiol Genomics 2007; 32: 28-32.
[http://dx.doi.org/10.1152/physiolgenomics.00165.2007]
[73]
Pease JC, Brewer M, Tirnauer JS. Spontaneous spheroid budding from monolayers: a potential contribution to ovarian cancer dissemination. Biol Open 2012; 1: 622-8.
[http://dx.doi.org/10.1242/bio.2012653]
[74]
Rubin BP, Duensing A. Mechanisms of resistance to small molecule kinase inhibition in the treatment of solid tumors. Lab Invest 2006; 86: 981-6.
[http://dx.doi.org/10.1038/labinvest.3700466]
[75]
Raguz S, Yagüe E. Resistance to chemotherapy: new treatments and novel insights into an old problem. Br J Cancer 2008; 99: 387-91.
[http://dx.doi.org/10.1038/sj.bjc.6604510]
[76]
Rao W, Zhao S, Yu J, Lu X, Zynger DL, He X. Enhanced enrichment of prostate cancer stem-like cells with miniaturized 3D culture in liquid core-hydrogel shell microcapsules. Biomaterials 2014; 35: 7762-73.
[http://dx.doi.org/10.1016/j.biomaterials.2014.06.011]
[77]
Lee J, Lilly GD, Doty RC, Podsiadlo P, Koto NA. In vitro toxicity testing of nanoparticles in 3D cell culture. Small 2009; 5: 1213-21.
[http://dx.doi.org/10.1002/smll.200801788]
[78]
Saleh FA, Genever PG. Turning round: multipotent stromal cells, a three-dimensional revolution? Cytotherapy 2011; 13: 903-12.
[http://dx.doi.org/10.3109/14653249.2011.586998]
[79]
Sha H, Zou Z, Xin K, et al. Tumor-penetrating peptide fused EGFR single-domain antibody enhances cancer drug penetration into 3D multicellular spheroids and facilitates effective gastric cancer therapy. J Control Release 2015; 200: 188-200.
[http://dx.doi.org/10.1016/j.jconrel.2014.12.039]
[80]
Sullivan R, Paré GC, Frederiksen LJ, Semenza GL, Graham CH. Hypoxia-induced resistance to anticancer drugs is associated with decreased senescence and requires hypoxia-inducible factor-1 activity. Mol Cancer Ther 2008; 7: 1961-73.
[http://dx.doi.org/10.1158/1535-7163.MCT-08-0198]
[81]
Shehzad A, Qureshi M, Jabeen S, et al. Synthesis, characterization and antibacterial activity of silver nanoparticles using Rhazya stricta. PeerJ 2018; 6e6086
[http://dx.doi.org/10.7717/peerj.6086]
[82]
Smith SJ, Wilson M, Ward JH, et al. Recapitulation of tumor heterogeneity and molecular signatures in a 3D brain cancer model with decreased sensitivity to histone deacetylase inhibition. PLoS One 2012; 7e52335
[http://dx.doi.org/10.1371/journal.pone.0052335]
[83]
Tibbitt MW, Anseth KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol Bioeng 2009; 103: 655-63.
[http://dx.doi.org/10.1002/bit.22361]
[84]
Van der Worp HB, Howells DW, Sena ES, et al. Can animal models of disease reliably inform human studies? PLoS Med 2010; 7e1000245
[http://dx.doi.org/10.1371/journal.pmed.1000245]
[85]
Vesuna F, Lisok A, Kimble B, Raman V. Twist modulates breast cancer stem cells by transcriptional regulation of CD24 expression. Neoplasia 2009; 11: 1318-28.
[http://dx.doi.org/10.1593/neo.91084]
[86]
Wenzel C, Riefke B, Gründemann S, et al. 3D high-content screening for the identification of compounds that target cells in dormant tumor spheroid regions. Exp Cell Res 2014; 323: 131-43.
[http://dx.doi.org/10.1016/j.yexcr.2014.01.017]
[87]
Weigelt B, Lo AT, Park CC, Gray JW, Bissell MJ. HER2 signaling pathway activation and response of breast cancer cells to HER2-targeting agents is dependent strongly on the 3D microenvironment. Breast Cancer Res Treat 2010; 122: 35-43.
[http://dx.doi.org/10.1007/s10549-009-0502-2]
[88]
Xu XX, Liu C, Liu Y, et al. Enrichment of cancer stem cell-like cells by culture in alginate gel beads. J Biotechnol 2014; 177: 1-12.
[http://dx.doi.org/10.1016/j.jbiotec.2014.02.016]
[89]
Yang Z, Zhao X. A 3D model of ovarian cancer cell lines on peptide nanofiber scaffold to explore the cell-scaffold interaction and chemotherapeutic resistance of anticancer drugs. Int J Nanomedicine 2011; 6: 303-10.
[http://dx.doi.org/10.2147/IJN.S15279]
[90]
Yip D, Cho CH. A multicellular 3D heterospheroid model of liver tumor and stromal cells in collagen gel for anti-cancer drug testing. Biochem Biophys Res Commun 2013; 433: 327-32.
[91]
Zhu L, Wang T, Perche F, Taigind A, Torchilin VP. Enhanced anticancer activity of nanopreparation containing an MMP2-sensitive PEG-drug conjugate and cell-penetrating moiety. Proc Natl Acad Sci USA 2013; 110: 17047-52.
[92]
Zschenker O, Streichert T, Hehlgans S, Cordes N. Genome-wide gene expression analysis in cancer cells reveals 3D growth to affect ECM and processes associated with cell adhesion but not DNA repair. PLoS One 2012; 7e34279
[http://dx.doi.org/10.1371/journal.pone.0034279]
[93]
Bouchet BP, Akhmanova A. Microtubules in 3D cell motility. J Cell Sci 2017; 130: 39-50.
[http://dx.doi.org/10.1242/jcs.189431]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy