2,949
Views
6
CrossRef citations to date
0
Altmetric
Review Articles

Advanced bioengineering technologies for preclinical research

, &
Article: 1622451 | Received 27 Mar 2019, Accepted 03 May 2019, Published online: 25 Jun 2019

References

  • Duval K, Grover H, Han L-H, et al. Modeling physiological events in 2D vs. 3D cell culture. Physiology. 2017;32:423–450.
  • Kacy Cullen D, Wolf JA, Vernekar VN, et al. Neural tissue engineering and biohybridized microsystems for neurobiological investigation in vitro (part 1). Crit Rev Biomed Eng. 2011;39:201–240.
  • Kimura H, Sakai Y, Fujii T. Organ/body-on-a-chip based on microfluidic technology for drug discovery. Drug Metab Pharmacokinet. 2018;33:43–48.
  • Lee SH, Sung JH. Organ-on-a-chip technology for reproducing multiorgan physiology. Adv Healthcare Mater. 2018;7:1700419.
  • Yamada KM, Cukierman E. Modeling tissue morphogenesis and cancer in 3D. Cell. 2007;130:601–610.
  • Hoarau-Véchot J, Rafii A, Touboul C, et al. Halfway between 2D and animal models: are 3D cultures the ideal tool to study cancer-microenvironment interactions? IJMS. 2018;19:181.
  • Edmondson R, Broglie JJ, Adcock AF, et al. Three-dimensional cell culture systems and their applications in drug discovery and cell-based biosensors. Assay Drug Dev Technol. 2014;12:207–218.
  • Shanti A, Teo J, Stefanini C. In vitro immune organs-on-chip for drug development: a review. Pharmaceutics. 2018;10:278.
  • Ericsson AC, Crim MJ, Franklin CL. A brief history of animal modeling. Mo Med. 2013;110:201–205.
  • Conn PM. source book of models for biomedical research. Totowa, NJ: Humana Press; 2008.
  • Seok J, Warren HS, Cuenca AG, et al. Genomic responses in mouse models poorly mimic human inflammatory diseases. Natl Acad Sci. 2013;110:3507–3512.
  • Polini A, Del Mercato LL, Barra A, et al. Towards the development of human immune-system-on-a-chip platforms. Drug Discov Today. 2018;24:1–9.
  • Lancaster MA, Knoblich JA. Organogenesis in a dish: modeling development and disease using organoid technologies. Science. 2014;345:1247125.
  • Rossi G, Manfrin A, Lutolf MP. Progress and potential in organoid research. Nat Rev Genet. 2018;19:1–17.
  • Eiraku M, Takata N, Ishibashi H, et al. Self-organizing optic-cup morphogenesis in three-dimensional culture. Nature. 2011;472:51–56.
  • Sato T, Vries RG, Snippert HJ, et al. Single Lgr5 stem cells build crypt-villus structures in vitro without a mesenchymal niche. Nature. 2009;459:262–265.
  • Takahashi T. Organoids for drug discovery and personalized medicine. Ann Rev Pharmacol Toxicol. 2019;59:447–462.
  • Lancaster MA, Renner M, Martin C-A, et al. Cerebral organoids model human brain development and microcephaly. Nature. 2013;501:373–379.
  • Huch M, Knoblich JA, Lutolf MP, et al. The hope and the hype of organoid research. Development. 2017;144:938–941.
  • Fang Y, Eglen RM. Three-dimensional cell cultures in drug discovery and Development. SLAS Discov. 2017;22:456–472.
  • Unger C, Kramer N, Walzl A, et al. Modeling human carcinomas: physiologically relevant 3D models to improve anti-cancer drug development. Adv Drug Deliv Rev. 2014;79:50–67.
  • Paul Weiss ACT. Reconstitution of complete organs from single-cell suspensions of chick embryos in advanced stages of differentiation. Proc Natl Acad Sci USA. 1960;46:1177–1185.
  • Boyle J, Luan B, Cruz TF, et al. Characterization of proteoglycan accumulation during formation of cartilagenous tissue in vitro. Osteoarthritis Cartilage. 1995;3:117–125.
  • Kandel RA, Boyle J, Gibson G, et al. In vitro formation of mineralized cartilagenous tissue by articular chondrocytes. In Vitro Cell Dev Biol Anim. 1997;33:174–181.
  • St-Pierre J-P, Gan L, Wang J, et al. The incorporation of a zone of calcified cartilage improves the interfacial shear strength between in vitro-formed cartilage and the underlying substrate. Acta Biomater. 2012;8:1603–1615.
  • St-Pierre J-P, Wang Q, Li SQ, et al. Inorganic polyphosphate stimulates cartilage tissue formation. Tissue Eng Part A. 2012;18:1282–1292.
  • De Croos JNA, Dhaliwal SS, Grynpas MD, et al. Cyclic compressive mechanical stimulation induces sequential catabolic and anabolic gene changes in chondrocytes resulting in increased extracellular matrix accumulation. Matrix Biol. 2006;25:323–331.
  • Kandel RA, Grynpas M, Pilliar R, et al. Repair of osteochondral defects with biphasic cartilage-calcium polyphosphate constructs in a sheep model. Biomaterials. 2006;27:4120–4131.
  • Sung JH, Yu J, Luo D, et al. Microscale 3-D hydrogel scaffold for biomimetic gastrointestinal (GI) tract model. Lab Chip. 2011;11:389–392.
  • Huang G, Wang L, Wang S, et al. Engineering three-dimensional cell mechanical microenvironment with hydrogels. Biofabrication. 2012;4:042001.
  • Tibbitt MW, Anseth KS. Hydrogels as extracellular matrix mimics for 3D cell culture. Biotechnol Bioeng. 2009;103:655–663.
  • DeForest CA, Anseth KS. Advances in bioactive hydrogels to probe and direct cell fate. Annu Rev Chem Biomol Eng. 2012;3:421–444.
  • Brown GE, Khetani SR. Microfabrication of liver and heart tissues for drug development. Philos Trans R Soc Lond B Biol Sci. 2018;373:20170225.
  • Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2007;26:120–126.
  • Ware BR, Berger DR, Khetani SR. Prediction of drug-induced liver injury in micropatterned co-cultures containing iPSC-derived human hepatocytes. Toxicol Sci. 2015;145:252–262.
  • Berger DR, Ware BR, Davidson MD, et al. Enhancing the functional maturity of induced pluripotent stem cell-derived human hepatocytes by controlled presentation of cell-cell interactions in vitro. Hepatology. 2015;61:1370–1381.
  • Radisic M, Park H, Shing H, Consi T, Schoen F, Langer R, Freed LE, Vunjak-Novakovic G. Functional assembly of engineered myocardium by electrical stimulation of cardiac myocytes cultured on scaffolds, Proc Natl Acad Sci USA 2004;101:18129–18134.
  • Tulloch NL, Muskheli V, Razumova MV, Korte FS, Regnier M, Hauch KD, Pabon L, Reinecke H, Murry CE. Growth of engineered human myocardium with mechanical loading and vascular coculture, Circ Res 2011;109:47–59.
  • Zhang D, Shadrin IY, Lam J, et al. Tissue-engineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials. 2013;34:5813–5820.
  • Turnbull IC, Karakikes I, Serrao GW, et al. Advancing functional engineered cardiac tissues toward a preclinical model of human myocardium. Faseb J. 2014;28:644–654.
  • Legant WR, Pathak A, Yang MT, et al. Microfabricated tissue gauges to measure and manipulate forces from 3D microtissues. Proc Natl Acad Sci USA. 2009;106:10097–10102.
  • Thavandiran N, Dubois N, Mikryukov A, et al. Design and formulation of functional pluripotent stem cell-derived cardiac microtissues. Proc Natl Acad Sci USA. 2013;110:E4698–707.
  • Tiburcy M, Hudson JE, Balfanz P, et al. Defined engineered human myocardium with advanced maturation for applications in heart failure modeling and repair. Circulation. 2017;135:1832–1847.
  • Nunes SS, Miklas JW, Liu J, et al. Biowire: a platform for maturation of human pluripotent stem cell-derived cardiomyocytes. Nat Methods. 2013;10:781–787.
  • Xiao Y, Zhang B, Liu H, et al. Microfabricated perfusable cardiac biowire: a platform that mimics native cardiac bundle. Lab Chip. 2014;14:869–882.
  • Amano Y, Nishiguchi A, Matsusaki M, et al. Development of vascularized iPSC derived 3D-cardiomyocyte tissues by filtration layer-by-layer technique and their application for pharmaceutical assays. Acta Biomater. 2016;33:110–121.
  • Mathur A, Loskill P, Shao K, et al. Human iPSC-based cardiac microphysiological system for drug screening applications. Sci Rep. 2015;5:8883.
  • Abaci HE, Coffman A, Doucet Y, et al. Tissue engineering of human hair follicles using a biomimetic developmental approach. Nat Commun. 2018;9:5301.
  • Vioux-Chagnoleau C, Lejeune F, Sok J, et al. Reconstructed human skin: from photodamage to sunscreen photoprotection and anti-aging molecules. J Dermatological Sci Suppl. 2006;2:S1–S12.
  • Zhang Z, Michniak-Kohn BB. Tissue engineered human skin equivalents. Pharmaceutics. 2012;4:26–41.
  • Feng Y, Mitchison TJ, Bender A, et al. Multi-parameter phenotypic profiling: using cellular effects to characterize small-molecule compounds. Nat Rev Drug Discov. 2009;8:567–578.
  • Asmani M, Velumani S, Li Y, et al. Fibrotic microtissue array to predict anti-fibrosis drug efficacy. Nat Commun. 2018;9:2066.
  • Ma Z, Huebsch N, Koo S, et al. Contractile deficits in engineered cardiac microtissues as a result of MYBPC3 deficiency and mechanical overload. Nat Biomed Eng. 2018;2:955–967.
  • Maffioletti SM, Sarcar S, Henderson ABH, et al. Three-dimensional human iPSC-derived artificial skeletal muscles model muscular dystrophies and enable multilineage tissue engineering. Cell Rep. 2018;23:899–908.
  • Chen Y, Lin Y, Davis KM, et al. Robust bioengineered 3D functional human intestinal epithelium. Sci Rep. 2015;13708.
  • DeCicco RePass MA, Chen Y, Lin Y, et al. Novel Bioengineered Three-Dimensional Human Intestinal Model for Long-Term Infection of Cryptosporidium parvum. Infect Immun. 2017;85.
  • Nguyen Hoang AT, Chen P, Juarez J, et al. Dendritic cell functional properties in a three-dimensional tissue model of human lung mucosa. Am J Physiol Lung Cell Mol Physiol. 2012;302:L226–37.
  • Mairpady Shambat S, Chen P, Nguyen Hoang AT, et al. Modelling staphylococcal pneumonia in a human 3D lung tissue model system delineates toxin-mediated pathology. Dis Model Mech. 2015;8:1413–1425.
  • Murphy SV, Atala A. 3D bioprinting of tissues and organs. Nature Biotechnology. 2014;32:773–785.
  • Pfister A, Landers R, Laib A, et al. Biofunctional rapid prototyping for tissue-engineering applications: 3D bioplotting versus 3D printing. J Polym Sci Part a-Polym Chem. 2004;42:624–638.
  • Ozbolat IT, Hospodiuk M. Current advances and future perspectives in extrusion-based bioprinting. Biomaterials. 2016;76:321–343.
  • Klebe RJ. Cytoscribing - a method for micropositioning cells and the construction of two-dimensional and 3-dimensional synthetic tissues. Exp Cell Res. 1988;179:362–373.
  • Gudapati H, Dey M, Ozbolat I. A comprehensive review on droplet-based bioprinting: past, present and future. Biomaterials. 2016;102:20–42.
  • Guillotin B, Souquet A, Catros S, et al. Laser assisted bioprinting of engineered tissue with high cell density and microscale organization. Biomaterials. 2010;31:7250–7256.
  • Lu Y, Mapili G, Suhali G, et al. A digital micro-mirror device-based system for the microfabrication of complex, spatially patterned tissue engineering scaffolds. J Biomed Mater Res A. 2006;77:396–405.
  • Ng WL, Qi JTZ, Yeong WY, et al. Proof-of-concept: 3D bioprinting of pigmented human skin constructs. Biofabrication. 2018;10:025005.
  • Ma X, Qu X, Zhu W, et al. Deterministically patterned biomimetic human iPSC-derived hepatic model via rapid 3D bioprinting. Proc Natl Acad Sci USA. 2016;113:2206–2211.
  • Carmeliet P, Jain RK. Angiogenesis in cancer and other diseases. Nature. 2000;407:249–257.
  • Rouwkema J, Khademhosseini A. Vascularization and angiogenesis in tissue engineering: beyond creating static networks. Trends Biotechnol. 2016;34:733–745.
  • Butt OI, Carruth R, Kutala VK, et al. Stimulation of peri-implant vascularization with bone marrow-derived progenitor cells: monitoring by in VivoEPR oximetry. Tissue Eng. 2007;13:2053–2061.
  • Zhang Y, Yu Y, Chen H, et al. Characterization of printable cellular micro-fluidic channels for tissue engineering. Biofabrication. 2013;5:025004.
  • Attalla R, Ling C, Selvaganapathy P. Fabrication and characterization of gels with integrated channels using 3D printing with microfluidic nozzle for tissue engineering applications. Biomed Microdevices. 2016;18:1241.
  • Datta P, Ayan B, Ozbolat IT. Bioprinting for vascular and vascularized tissue biofabrication. Acta Biomater. 2017;51:1–20.
  • Kolesky DB, Homan KA, Skylar-Scott MA, et al. Three-dimensional bioprinting of thick vascularized tissues. Proc Natl Acad Sci USA. 2016;113:3179–3184.
  • Miller JS, Stevens KR, Yang MT, et al. Rapid casting of patterned vascular networks for perfusable engineered three-dimensional tissues. Nat Mater. 2012;11:768–74.
  • Miri AK, Khalilpour A, Cecen B, et al. Multiscale bioprinting of vascularized models. Biomaterials. 2019;198:204–216.
  • Parak A, Pradeep P, Du Toit LC, et al. Functionalizing bioinks for 3D bioprinting applications. Drug Discov Today. 2019;24:198–205.
  • Ma X, Liu J, Zhu W, et al. 3D bioprinting of functional tissue models for personalized drug screening and in vitro disease modeling. Adv Drug Deliv Rev. 2018;132:235–251.
  • Combellack E, Jessop ZM, Whitaker IS. The commercial 3D bioprinting industry. New York: Elsevier Ltd; 2018.
  • Datta P, Barui A, Wu Y, et al. Essential steps in bioprinting from pre- to post-bioprinting. Biotechnol Adv. 2018;36:1481–1504.
  • Blundell C, Tess ER, Schanzer ASR, et al. A microphysiological model of the human placental barrier. Lab Chip. 2016;16:3065–3073.
  • Jackson EL, Lu H. Three-dimensional models for studying development and disease: moving on from organisms to organs-on-a-chip and organoids. Integr Biol. 2016;8:672–683.
  • Ahadian S, Civitarese R, Bannerman D, et al. Organ-on-a-chip platforms: a convergence of advanced materials, cells, and microscale technologies. Adv Healthcare Mater. 2017;7:1700506–1700553.
  • Mittal R, Woo FW, Castro CS, et al. Organ-on-chip models: implications in drug discovery and clinical applications. J Cell Physiol. 2018;105:e48–29.
  • Rothbauer M, Rosser JM, Zirath H, et al. ScienceDirect tomorrow today: organ-on-a-chip advances towards clinically relevant pharmaceutical and medical in vitro models. Curr Opin Biotechnol. 2018;55:81–86.
  • Huh D, Matthews BD, Mammoto A, et al. Reconstituting organ-level lung functions on a chip. Science. 2010;328:1658–1662.
  • Nawroth JC, Barrile R, Conegliano D, et al. Stem cell-based lung-on-chips: the best of both worlds? Adv Drug Deliv Rev. 2018;1–21.
  • Ronaldson-Bouchard K, Vunjak-Novakovic G. Organs-on-a-chip: a fast track for Engineered human tissues in drug development. Stem Cell. 2018;22:310–324.
  • Esch EW, Bahinski A, Huh D. Organs-on-chips at the frontiers of drug discovery. Nat Rev Drug Discov. 2015;14:248–260.
  • Shi Y, Inoue H, Wu JC, et al. Induced pluripotent stem cell technology: a decade of progress. Nat Publishing Group. 2016;16:115–130.
  • van Den Berg A, Mummery CL, Passier R, et al. Personalised organs-on-chips: functional testing for precision medicine. Lab Chip. 2019;19:198–205.
  • Sosa-Hernández JE, Villalba-Rodríguez AM, Romero-Castillo KD, et al. Organs-on-a-chip module: a review from the development and applications perspective. Micromachines. 2018;9:536.
  • Zhang B, Korolj A, Lai BFL, et al. Advances in organ-on-a-chip engineering. Nat Rev Mater. 2018;3:1–22.
  • Sun W, Luo Z, Lee J, et al. Organ-on-a-chip for cancer and immune organs modeling. Adv Healthcare Mater. 2019;177:1801363.
  • Nawroth J, Rogal J, Weiss M, et al. Organ-on-a-chip systems for women’s health applications. Adv Healthcare Mater. 2017;7:1700550.
  • Low LA, Tagle DA. Tissue chips – innovative tools for drug development and disease modeling. Lab Chip. 2017;17:3026–3036.
  • Ortega-Prieto AM, Skelton JK, Wai SN, et al. 3D microfluidic liver cultures as a physiological preclinical tool for hepatitis B virus infection. Nat Commun. 2018;9:1–15.
  • Ahn J, Ko J, Lee S, et al. Microfluidics in nanoparticle drug delivery; From synthesis to pre-clinical screening. Adv Drug Deliv Rev. 2018;128:29–53.
  • Aziz A, Geng C, Fu M, et al. The role of microfluidics for organ on chip simulations. Bioengineering. 2017;4:39.
  • Viswanathan P, Guvendiren M, Chua W, et al. Mimicking the topography of the epidermal-dermal interface with elastomer substrates. Integr Biol. 2016;8:21–29.
  • Kageyama T, Yoshimura C, Myasnikova D, et al. Spontaneous hair follicle germ (HFG) formation in vitro, enabling the large-scale production of HFGs for regenerative medicine. Biomaterials. 2018;154:291–300.
  • Ortega I, Ryan AJ, Deshpande P, et al. Combined microfabrication and electrospinning to produce 3-D architectures for corneal repair. Acta Biomater. 2013;9:5511–5520.
  • Levis HJ, Massie I, Dziasko MA, et al. Rapid tissue engineering of biomimetic human corneal limbal crypts with 3D niche architecture. Biomaterials. 2013;34:8860–8868.
  • Subramanian B, Kaya O, Pollak MR, et al. Guided tissue organization and disease modeling in a kidney tubule array. Biomaterials. 2018;183:295–305.
  • Wang YI, Carmona C, Hickman JJ, et al. Multiorgan microphysiological systems for drug development: strategies, advances, and challenges. Adv Healthcare Mater. 2018;7:1701000.
  • Castaño AG, García-Díaz M, Torras N, et al. Dynamic photopolymerization produces complex microstructures on hydrogels in a moldless approach to generate a 3D intestinal tissue model. Biofabrication. 2019;11:1–16.
  • Zhang YS, Aleman J, Shin SR, et al. Multisensor-integrated organs-on-chips platform for automated and continual in situ monitoring of organoid behaviors. Proc Natl Acad Sci USA. 2017;114:E2293–E2302.
  • Zervantonakis IK, Hughes-Alford SK, Charest JL, et al. Three-dimensional microfluidic model for tumor cell intravasation and endothelial barrier function. Proc Natl Acad Sci USA. 2012;109:13515–13520.
  • Das M, Wilson K, Molnar P, et al. Differentiation of skeletal muscle and integration of myotubes with silicon microstructures using serum-free medium and a synthetic silane substrate. Nat Protoc. 2007;2:1795–1801.
  • Varghese K, Molnar P, Das M, et al. A new target for amyloid beta toxicity validated by standard and high-throughput electrophysiology. Plos One. 2010;5:e8643.
  • Walter FR, Valkai S, Kincses A, et al. A versatile lab-on-a-chip tool for modeling biological barriers. Sens Actuators B Chem. 2016;222:1209–1219.
  • Takebe T, Zhang B, Radisic M. Synergistic engineering: organoids meet organs-on-a-chip. Cell Stem Cell. 2017;21:297–300.
  • Zhao H, Chen Y, Shao L, et al. Airflow‐assisted 3D bioprinting of human heterogeneous microspheroidal organoids with microfluidic nozzle. Small. 2018;14:1802630–1802637.
  • Peng W, Datta P, Wu Y, et al. Challenges in bio-fabrication of organoid cultures. Cell Biol Transl Med. 2018;3:53–71. Advances in Experimental Medicine and Biology, vol 1107. Springer, Cham.