Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip
Abstract
:1. Introduction
2. BioMEMS
3. Microfluidics
4. Lab-on-a-Chip
Lab-on-a-Chip Devices for Point-of-Care Diagnostics
5. Organ-on-a-Chip
5.1. Current Organ-on-a-Chip Platforms
5.1.1. Lung
5.1.2. Cardiovascular
5.1.3. Brain
5.1.4. Liver
5.1.5. Kidney
5.1.6. Gut
5.1.7. Skin
5.2. Body-on-a-Chip
5.3. Organ-on-a-Chip Market
6. Discussion and Future Perspectives
7. Conclusions
Funding
Acknowledgments
Conflicts of Interest
List of Acronyms
BioMEMS | Biomedical Micro-Electro-Mechanical Systems |
CE | Capillary Electrophoresis |
CMs | Cardiomyocytes |
CROs | Contract research organizations |
ECM | Extracellular Matrix |
GC | Gas Cromatography |
GPC | Gas-phase chromatography |
hCG | human Chorionic Gonadortropin |
HPLC | High-Pressure Liquid Chromatography |
hPSCs | human Pluripotent Stem Cells |
iPSCs | Induced-pluripotent stem cells |
IC | Integrated Circuits |
IL | Interleukin |
LOC | Lab-on-a-chip |
MEMS | Micro-Electro-Mechanical Systems |
OOC | Organ-on-a-chip |
PBPK | Pharmacokinetics |
PD | Pharmacodynamics |
PDMS | Polydimethylsiloxane |
POC | Point-of-Care |
PTFE | Polytetrafluoroethylene |
R&D | Research and development |
UV | Ultraviolet |
μTAS | Micro-Total Analysis Systems |
References
- Grayson, A.C.R.; Shawgo, R.S.; Johnson, A.M.; Flynn, N.T.; Li, Y.; Cima, M.J.; Langer, R. A BioMEMS review: MEMS technology for physiologically integrated devices. Proc. IEEE 2004, 92, 6–21. [Google Scholar] [CrossRef]
- Verpoorte, E.; De Rooij, N.F. Microfluidics meets MEMS. Proc. IEEE 2003, 91, 930–953. [Google Scholar] [CrossRef] [Green Version]
- Convery, N.; Gadegaard, N. 30 years of microfluidics. Micro Nano Eng. 2019, 2, 76–91. [Google Scholar] [CrossRef]
- Whitesides, G.M. The origins and the future of microfluidics. Nature 2006, 442, 368–373. [Google Scholar] [CrossRef]
- Astolfi, M.; Péant, B.; Lateef, M.; Rousset, N.; Kendall-Dupont, J.; Carmona, E.; Monet, F.; Saad, F.; Provencher, D.; Mes-Masson, A.-M. Micro-dissected tumor tissues on chip: An ex vivo method for drug testing and personalized therapy. Lab Chip 2016, 16, 312–325. [Google Scholar] [CrossRef]
- Bhatia, S.N.; Ingber, D.E. Microfluidic organs-on-chips. Nat. Biotechnol. 2014, 32, 760–772. [Google Scholar] [CrossRef]
- Huh, D.; Hamilton, G.A.; Ingber, D.E. From 3D cell culture to organs-on-chips. Trends Cell Biol. 2011, 21, 745–754. [Google Scholar] [CrossRef] [Green Version]
- Sun, W.; Luo, Z.; Lee, J.; Kim, H.J.; Lee, K.; Tebon, P.; Feng, Y.; Dokmeci, M.R.; Sengupta, S.; Khademhosseini, A. Organ-on-a-Chip for Cancer and Immune Organs Modeling. Adv. Healthc. Mater. 2019, 8, 1801363. [Google Scholar] [CrossRef]
- Ahadian, S.; Civitarese, R.; Bannerman, D.; Mohammadi, M.H.; Lu, R.; Wang, E.; Davenport-Huyer, L.; Lai, B.; Zhang, B.; Zhao, Y. Organ-On-A-Chip Platforms: A Convergence of Advanced Materials, Cells, and Microscale Technologies. Adv. Healthc. Mater. 2018, 7, 1700506. [Google Scholar] [CrossRef]
- Bhise, N.S.; Ribas, J.; Manoharan, V.; Zhang, Y.S.; Polini, A.; Massa, S.; Dokmeci, M.R.; Khademhosseini, A. Organ-on-a-chip platforms for studying drug delivery systems. J. Control. Release 2014, 190, 82–93. [Google Scholar] [CrossRef] [Green Version]
- Polini, A.; Prodanov, L.; Bhise, N.S.; Manoharan, V.; Dokmeci, M.R.; Khademhosseini, A. Organs-on-a-chip: A new tool for drug discovery. Expert Opin. Drug Discov. 2014, 9, 335–352. [Google Scholar] [CrossRef] [PubMed]
- Huh, D.; Matthews, B.D.; Mammoto, A.; Montoya-Zavala, M.; Hsin, H.Y.; Ingber, D.E. Reconstituting organ-level lung functions on a chip. Science 2010, 328, 1662–1668. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.S.; Zhang, Y.-N.; Zhang, W. Cancer-on-a-chip systems at the frontier of nanomedicine. Drug Discov. Today 2017, 22, 1392–1399. [Google Scholar] [CrossRef]
- Huh, D.; Leslie, D.C.; Matthews, B.D.; Fraser, J.P.; Jurek, S.; Hamilton, G.A.; Thorneloe, K.S.; McAlexander, M.A.; Ingber, D.E. A human disease model of drug toxicity–induced pulmonary edema in a lung-on-a-chip microdevice. Sci. Transl. Med. 2012, 4, ra147–ra159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, W.; Sun, M.; Lu, B.; Yan, M.; Han, K.; Wang, J. A microfluidic platform for multi-size 3D tumor culture, monitoring and drug resistance testing. Sens. Actuators B Chem. 2019, 292, 111–120. [Google Scholar] [CrossRef]
- Huh, D.; Kim, H.J.; Fraser, J.P.; Shea, D.E.; Khan, M.; Bahinski, A.; Hamilton, G.A.; Ingber, D.E. Microfabrication of human organs-on-chips. Nat. Protoc. 2013, 8, 2135–2157. [Google Scholar] [CrossRef]
- Bhatia, S.N.; Chen, C.S. Tissue engineering at the micro-scale. Biomed. Microdevices 1999, 2, 131–144. [Google Scholar] [CrossRef]
- Chin, C.D.; Linder, V.; Sia, S.K. Lab-on-a-chip devices for global health: Past studies and future opportunities. Lab Chip 2007, 7, 41–57. [Google Scholar] [CrossRef]
- Abgrall, P.; Gue, A. Lab-on-chip technologies: Making a microfluidic network and coupling it into a complete microsystem—A review. J. Micromech. Microeng. 2007, 17, R15. [Google Scholar] [CrossRef]
- Temiz, Y.; Lovchik, R.D.; Kaigala, G.V.; Delamarche, E. Lab-on-a-chip devices: How to close and plug the lab? Microelectron. Eng. 2015, 132, 156–175. [Google Scholar] [CrossRef]
- Kovacs, G.T.; Maluf, N.I.; Petersen, K.E. Bulk micromachining of silicon. Proc. IEEE 1998, 86, 1536–1551. [Google Scholar] [CrossRef] [Green Version]
- Bustillo, J.M.; Howe, R.T.; Muller, R.S. Surface micromachining for microelectromechanical systems. Proc. IEEE 1998, 86, 1552–1574. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, M.A. Wafer-to-wafer bonding for microstructure formation. Proc. IEEE 1998, 86, 1575–1585. [Google Scholar] [CrossRef]
- Manz, A.; Graber, N.; Widmer, H.Á. Miniaturized total chemical analysis systems: A novel concept for chemical sensing. Sens. Actuators B Chem. 1990, 1, 244–248. [Google Scholar] [CrossRef]
- Manz, A.; Harrison, D.J.; Verpoorte, E.M.; Fettinger, J.C.; Paulus, A.; Lüdi, H.; Widmer, H.M. Planar chips technology for miniaturization and integration of separation techniques into monitoring systems: Capillary electrophoresis on a chip. J. Chromatogr. A 1992, 593, 253–258. [Google Scholar] [CrossRef]
- Jacobson, S.C.; Hergenroder, R.; Koutny, L.B.; Ramsey, J.M. High-speed separations on a microchip. Anal. Chem. 1994, 66, 1114–1118. [Google Scholar] [CrossRef]
- Harrison, D.J.; Fluri, K.; Seiler, K.; Fan, Z.; Effenhauser, C.S.; Manz, A. Micromachining a miniaturized capillary electrophoresis-based chemical analysis system on a chip. Science 1993, 261, 895–897. [Google Scholar] [CrossRef]
- Puleo, C.M.; Yeh, H.-C.; Wang, T.-H. Applications of MEMS technologies in tissue engineering. Tissue Eng. 2007, 13, 2839–2854. [Google Scholar] [CrossRef]
- Bashir, R. BioMEMS: State-of-the-art in detection, opportunities and prospects. Adv. Drug Deliv. Rev. 2004, 56, 1565–1586. [Google Scholar] [CrossRef]
- Polla, D.L.; Erdman, A.G.; Robbins, W.P.; Markus, D.T.; Diaz-Diaz, J.; Rizq, R.; Nam, Y.; Brickner, H.T.; Wang, A.; Krulevitch, P. Microdevices in medicine. Annu. Rev. Biomed. Eng. 2000, 2, 551–576. [Google Scholar] [CrossRef]
- Dittrich, P.S.; Manz, A. Lab-on-a-chip: Microfluidics in drug discovery. Nat. Rev. Drug Discov. 2006, 5, 210. [Google Scholar] [CrossRef] [PubMed]
- Quake, S.R.; Scherer, A. From micro-to nanofabrication with soft materials. Science 2000, 290, 1536–1540. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, B.; Radisic, M. Organ-on-a-chip devices advance to market. Lab Chip 2017, 17, 2395–2420. [Google Scholar] [CrossRef] [PubMed]
- Tsao, C.-W. Polymer microfluidics: Simple, low-cost fabrication process bridging academic lab research to commercialized production. Micromachines 2016, 7, 225. [Google Scholar] [CrossRef] [Green Version]
- Becker, H.; Gärtner, C. Polymer microfabrication technologies for microfluidic systems. Anal. Bioanal. Chem. 2008, 390, 89–111. [Google Scholar] [CrossRef]
- Giboz, J.; Copponnex, T.; Mélé, P. Microinjection molding of thermoplastic polymers: A review. J. Micromech. Microeng. 2007, 17, R96. [Google Scholar] [CrossRef]
- Kim, B.J.; Meng, E. Review of polymer MEMS micromachining. J. Micromech. Microeng. 2015, 26, 013001. [Google Scholar] [CrossRef]
- Voldman, J. BioMEMS: Building with cells. Nat. Mater. 2003, 2, 433. [Google Scholar] [CrossRef]
- Mark, D.; Haeberle, S.; Roth, G.; Von Stetten, F.; Zengerle, R. Microfluidic Lab-On-a-Chip Platforms: Requirements, Characteristics and Applications. In Microfluidics Based Microsystems; Springer: Berlin/Heidelberg, Germany, 2010; pp. 305–376. [Google Scholar]
- Canny, M. Flow and transport in plants. Annu. Rev. Fluid Mech. 1977, 9, 275–296. [Google Scholar] [CrossRef]
- Chin, C.D.; Linder, V.; Sia, S.K. Commercialization of microfluidic point-of-care diagnostic devices. Lab Chip 2012, 12, 2118–2134. [Google Scholar] [CrossRef]
- Whitesides, G.M. What comes next? Lab Chip 2011, 11, 191–193. [Google Scholar] [CrossRef] [PubMed]
- Stone, H.A.; Stroock, A.D.; Ajdari, A. Engineering flows in small devices: Microfluidics toward a lab-on-a-chip. Annu. Rev. Fluid Mech. 2004, 36, 381–411. [Google Scholar] [CrossRef] [Green Version]
- Saliba, J.; Daou, A.; Damiati, S.; Saliba, J.; El-Sabban, M.; Mhanna, R. Development of microplatforms to mimic the in vivo architecture of CNS and PNS physiology and their diseases. Genes 2018, 9, 285. [Google Scholar] [CrossRef] [Green Version]
- Squires, T.M.; Quake, S.R. Microfluidics: Fluid physics at the nanoliter scale. Rev. Mod. Phys. 2005, 77, 977. [Google Scholar] [CrossRef] [Green Version]
- Petri, R. Eine kleine modification des Koch’schen plattenverfahrens. Cntralbl. Bakteriol. Pasitenkunde 1887, 1, 279–280. [Google Scholar]
- Woolley, A.T.; Mathies, R.A. Ultra-high-speed DNA fragment separations using microfabricated capillary array electrophoresis chips. Proc. Natl. Acad. Sci. USA 1994, 91, 11348–11352. [Google Scholar] [CrossRef] [Green Version]
- Brody, J.P.; Han, Y.; Austin, R.H.; Bitensky, M. Deformation and flow of red blood cells in a synthetic lattice: Evidence for an active cytoskeleton. Biophys. J. 1995, 68, 2224–2232. [Google Scholar] [CrossRef] [Green Version]
- Srinivasan, K.; Pohl, C.; Avdalovic, N. Cross-linked polymer coatings for capillary electrophoresis and application to analysis of basic proteins, acidic proteins, and inorganic ions. Anal. Chem. 1997, 69, 2798–2805. [Google Scholar] [CrossRef]
- Li, P.C.; Harrison, D.J. Transport, manipulation, and reaction of biological cells on-chip using electrokinetic effects. Anal. Chem. 1997, 69, 1564–1568. [Google Scholar] [CrossRef]
- Hessel, V.; Löwe, H.; Schönfeld, F. Micromixers—A review on passive and active mixing principles. Chem. Eng. Sci. 2005, 60, 2479–2501. [Google Scholar] [CrossRef]
- Daw, R.; Finkelstein, J. Insight: Lab on a chip. Nature 2006, 442, 367–418. [Google Scholar] [CrossRef] [Green Version]
- Lei, K.F. Introduction: The Origin, Current Status, and Future of Microfluidics. Microfluid. Fundam. Devices Appl. Fundam. Appl. 2018, 1–18. [Google Scholar] [CrossRef]
- Raje, P.V.; Murmu, N.C. A review on electrohydrodynamic-inkjet printing technology. Int. J. Emerg. Technol. Adv. Eng. 2014, 4, 174–183. [Google Scholar]
- Terry, S.C.; Jerman, J.H.; Angell, J.B. A gas chromatographic air analyzer fabricated on a silicon wafer. IEEE Trans. Electron Devices 1979, 26, 1880–1886. [Google Scholar] [CrossRef]
- Shoji, S.; Esashi, M.; Matsuo, T. Prototype miniature blood gas analyser fabricated on a silicon wafer. Sens. Actuators 1988, 14, 101–107. [Google Scholar] [CrossRef]
- Zengerle, R.; Ulrich, J.; Kluge, S.; Richter, M.; Richter, A. A bidirectional silicon micropump. Sens. Actuators A Phys. 1995, 50, 81–86. [Google Scholar] [CrossRef]
- Pacifici, R.; Farré, M.; Pichini, S.; Ortuño, J.; Roset, P.N.; Zuccaro, P.; Segura, J.; de la Torre, R. Sweat testing of MDMA with the Drugwipe® analytical device: A controlled study with two volunteers. J. Anal. Toxicol. 2001, 25, 144–146. [Google Scholar] [CrossRef]
- Hicks, J.; Iosefsohn, M. Reliability of home pregnancy-test kits in the hands of laypersons. N. Engl. J. Med. 1989, 320, 320–321. [Google Scholar]
- Wu, A.H. Theme: Immunoassay Automation at the Millennium-Laboratory and Near Patient Testing for Cardiac Markers. J. Clin. Ligand Assay 1999, 22, 32–37. [Google Scholar]
- Haeberle, S.; Zengerle, R. Microfluidic platforms for lab-on-a-chip applications. Lab Chip 2007, 7, 1094–1110. [Google Scholar] [CrossRef] [PubMed]
- Srinivasan, V.; Pamula, V.K.; Fair, R.B. An integrated digital microfluidic lab-on-a-chip for clinical diagnostics on human physiological fluids. Lab Chip 2004, 4, 310–315. [Google Scholar] [CrossRef] [PubMed]
- Nabovati, G.; Ghafar-Zadeh, E.; Letourneau, A.; Sawan, M. Towards High Throughput Cell Growth Screening: A New CMOS 8$\times $8 Biosensor Array for Life Science Applications. IEEE Trans. Biomed. Circuits Syst. 2016, 11, 380–391. [Google Scholar] [CrossRef] [PubMed]
- Ozhikandathil, J.; Badilescu, S.; Packirisamy, M. Gold nanoisland structures integrated in a lab-on-a-chip for plasmonic detection of bovine growth hormone. J. Biomed. Opt. 2012, 17, 077001. [Google Scholar] [CrossRef] [PubMed]
- Becker, H.; Hansen-Hagge, T.; Gärtner, C. Microfluidic Devices for Rapid Identification and Characterization of Pathogens. In Biological Identification; Elsevier: Amsterdam, The Netherlands, 2014; pp. 220–249. [Google Scholar]
- Jung, W.; Han, J.; Choi, J.-W.; Ahn, C.H. Point-of-care testing (POCT) diagnostic systems using microfluidic lab-on-a-chip technologies. Microelectron. Eng. 2015, 132, 46–57. [Google Scholar] [CrossRef]
- St John, A.; Price, C.P. Existing and emerging technologies for point-of-care testing. Clin. Biochem. Rev. 2014, 35, 155. [Google Scholar]
- Kozel, T.R.; Burnham-Marusich, A.R. Point-of-care testing for infectious diseases: Past, present, and future. J. Clin. Microbiol. 2017, 55, 2313–2320. [Google Scholar] [CrossRef] [Green Version]
- Schito, M.L.; Patricia D’Souza, M.; Michele Owen, S.; Busch, M.P. Challenges for rapid molecular HIV diagnostics. J. Infect. Dis. 2010, 201, S1–S6. [Google Scholar] [CrossRef]
- Weigl, B.; Domingo, G.; LaBarre, P.; Gerlach, J. Towards non-and minimally instrumented, microfluidics-based diagnostic devices. Lab Chip 2008, 8, 1999–2014. [Google Scholar] [CrossRef] [Green Version]
- Heller, A.; Feldman, B. Electrochemical glucose sensors and their applications in diabetes management. Chem. Rev. 2008, 108, 2482–2505. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.; Peng, C.; Jin, Z.; Xu, C. Development and evaluation of a rapid lateral flow immunochromatographic strip assay for screening 19-nortestosterone. Biomed. Chromatogr. 2007, 21, 861–866. [Google Scholar] [CrossRef]
- Leavitt, S.A. A private little revolution: The home pregnancy test in American culture. Bull. Hist. Med. 2006, 80, 317–345. [Google Scholar] [CrossRef] [PubMed]
- Posthuma-Trumpie, G.A.; Korf, J.; van Amerongen, A. Lateral flow (immuno) assay: Its strengths, weaknesses, opportunities and threats. A literature survey. Anal. Bioanal. Chem. 2009, 393, 569–582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, J. Electrochemical glucose biosensors. Chem. Rev. 2008, 108, 814–825. [Google Scholar] [CrossRef] [PubMed]
- Kuhn, L.S. Biosensors: Blockbuster or bomb? Electrochem. Soc. Interface 1998, 7, 26–31. [Google Scholar]
- Young, E.W. Cells, tissues, and organs on chips: Challenges and opportunities for the cancer tumor microenvironment. Integr. Biol. 2013, 5, 1096–1109. [Google Scholar] [CrossRef]
- Ahangar, P.; Cooke, M.E.; Weber, M.H.; Rosenzweig, D.H. Current biomedical applications of 3D printing and additive manufacturing. Appl. Sci. 2019, 9, 1713. [Google Scholar] [CrossRef] [Green Version]
- Huh, D.; Torisawa, Y.-S.; Hamilton, G.A.; Kim, H.J.; Ingber, D.E. Microengineered physiological biomimicry: Organs-on-chips. Lab Chip 2012, 12, 2156–2164. [Google Scholar] [CrossRef]
- Han, S.; Yan, J.-J.; Shin, Y.; Jeon, J.J.; Won, J.; Jeong, H.E.; Kamm, R.D.; Kim, Y.-J.; Chung, S. A versatile assay for monitoring in vivo-like transendothelial migration of neutrophils. Lab Chip 2012, 12, 3861–3865. [Google Scholar] [CrossRef]
- Sosa-Hernández, J.; Villalba-Rodríguez, A.; Romero-Castillo, K.; Aguilar-Aguila-Isaías, M.; García-Reyes, I.; Hernández-Antonio, A.; Ahmed, I.; Sharma, A.; Parra-Saldívar, R.; Iqbal, H. Organs-on-a-chip module: A review from the development and applications perspective. Micromachines 2018, 9, 536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Douville, N.J.; Zamankhan, P.; Tung, Y.-C.; Li, R.; Vaughan, B.L.; Tai, C.-F.; White, J.; Christensen, P.J.; Grotberg, J.B.; Takayama, S. Combination of fluid and solid mechanical stresses contribute to cell death and detachment in a microfluidic alveolar model. Lab Chip 2011, 11, 609–619. [Google Scholar] [CrossRef] [PubMed]
- Stucki, A.O.; Stucki, J.D.; Hall, S.R.; Felder, M.; Mermoud, Y.; Schmid, R.A.; Geiser, T.; Guenat, O.T. A lung-on-a-chip array with an integrated bio-inspired respiration mechanism. Lab Chip 2015, 15, 1302–1310. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, A.; Barrile, R.; van der Meer, A.D.; Mammoto, A.; Mammoto, T.; De Ceunynck, K.; Aisiku, O.; Otieno, M.A.; Louden, C.S.; Hamilton, G.A. Primary human lung alveolus-on-a-chip model of intravascular thrombosis for assessment of therapeutics. Clin. Pharmacol. Ther. 2018, 103, 332–340. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, T.; Duong Bang, D.; Wolff, A. 2019 novel coronavirus disease (COVID-19): Paving the road for rapid detection and point-of-care diagnostics. Micromachines 2020, 11, 306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Si, L.; Bai, H.; Rodas, M.; Cao, W.; Oh, C.Y.; Jiang, A.; Nurani, A.; Zhu, D.Y.; Goyal, G.; Gilpin, S. Human organs-on-chips as tools for repurposing approved drugs as potential influenza and COVID19 therapeutics in viral pandemics. bioRxiv 2020. [Google Scholar] [CrossRef]
- Shao, J.; Wu, L.; Wu, J.; Zheng, Y.; Zhao, H.; Jin, Q.; Zhao, J. Integrated microfluidic chip for endothelial cells culture and analysis exposed to a pulsatile and oscillatory shear stress. Lab Chip 2009, 9, 3118–3125. [Google Scholar] [CrossRef]
- Song, J.W.; Gu, W.; Futai, N.; Warner, K.A.; Nor, J.E.; Takayama, S. Computer-controlled microcirculatory support system for endothelial cell culture and shearing. Anal. Chem. 2005, 77, 3993–3999. [Google Scholar] [CrossRef]
- Eschenhagen, T.; Force, T.; Ewer, M.S.; De Keulenaer, G.W.; Suter, T.M.; Anker, S.D.; Avkiran, M.; De Azambuja, E.; Balligand, J.L.; Brutsaert, D.L. Cardiovascular side effects of cancer therapies: A position statement from the Heart Failure Association of the European Society of Cardiology. Eur. J. Heart Fail. 2011, 13, 1–10. [Google Scholar] [CrossRef]
- Pacher, P.; Kecskemeti, V. Cardiovascular side effects of new antidepressants and antipsychotics: New drugs, old concerns? Curr. Pharm. Des. 2004, 10, 2463–2475. [Google Scholar] [CrossRef] [Green Version]
- Au, H.T.H.; Cui, B.; Chu, Z.E.; Veres, T.; Radisic, M. Cell culture chips for simultaneous application of topographical and electrical cues enhance phenotype of cardiomyocytes. Lab Chip 2009, 9, 564–575. [Google Scholar]
- Marsano, A.; Conficconi, C.; Lemme, M.; Occhetta, P.; Gaudiello, E.; Votta, E.; Cerino, G.; Redaelli, A.; Rasponi, M. Beating heart on a chip: A novel microfluidic platform to generate functional 3D cardiac microtissues. Lab Chip 2016, 16, 599–610. [Google Scholar] [CrossRef]
- Ahn, S.; Ardoña, H.A.M.; Lind, J.U.; Eweje, F.; Kim, S.L.; Gonzalez, G.M.; Liu, Q.; Zimmerman, J.F.; Pyrgiotakis, G.; Zhang, Z. Mussel-inspired 3D fiber scaffolds for heart-on-a-chip toxicity studies of engineered nanomaterials. Anal. Bioanal. Chem. 2018, 410, 6141–6154. [Google Scholar] [CrossRef] [PubMed]
- Lind, J.U.; Busbee, T.A.; Valentine, A.D.; Pasqualini, F.S.; Yuan, H.; Yadid, M.; Park, S.-J.; Kotikian, A.; Nesmith, A.P.; Campbell, P.H. Instrumented cardiac microphysiological devices via multimaterial three-dimensional printing. Nat. Mater. 2017, 16, 303. [Google Scholar] [CrossRef]
- Xiao, Y.; Zhang, B.; Liu, H.; Miklas, J.W.; Gagliardi, M.; Pahnke, A.; Thavandiran, N.; Sun, Y.; Simmons, C.; Keller, G. Microfabricated perfusable cardiac biowire: A platform that mimics native cardiac bundle. Lab Chip 2014, 14, 869–882. [Google Scholar] [CrossRef]
- Ren, L.; Liu, W.; Wang, Y.; Wang, J.-C.; Tu, Q.; Xu, J.; Liu, R.; Shen, S.-F.; Wang, J. Investigation of hypoxia-induced myocardial injury dynamics in a tissue interface mimicking microfluidic device. Anal. Chem. 2012, 85, 235–244. [Google Scholar] [CrossRef] [PubMed]
- Günther, A.; Yasotharan, S.; Vagaon, A.; Lochovsky, C.; Pinto, S.; Yang, J.; Lau, C.; Voigtlaender-Bolz, J.; Bolz, S.-S. A microfluidic platform for probing small artery structure and function. Lab Chip 2010, 10, 2341–2349. [Google Scholar] [CrossRef] [PubMed]
- Xiong, Y.; Mahmood, A.; Chopp, M. Animal models of traumatic brain injury. Nat. Rev. Neurosci. 2013, 14, 128. [Google Scholar] [CrossRef] [Green Version]
- Caplin, J.D.; Granados, N.G.; James, M.R.; Montazami, R.; Hashemi, N. Microfluidic organ-on-a-chip technology for advancement of drug development and toxicology. Adv. Healthc. Mater. 2015, 4, 1426–1450. [Google Scholar] [CrossRef] [Green Version]
- Taylor, A.M.; Blurton-Jones, M.; Rhee, S.W.; Cribbs, D.H.; Cotman, C.W.; Jeon, N.L. A microfluidic culture platform for CNS axonal injury, regeneration and transport. Nat. Methods 2005, 2, 599. [Google Scholar] [CrossRef]
- Park, J.; Kim, S.; Park, S.I.; Choe, Y.; Li, J.; Han, A. A microchip for quantitative analysis of CNS axon growth under localized biomolecular treatments. J. Neurosci. Methods 2014, 221, 166–174. [Google Scholar] [CrossRef] [Green Version]
- Park, J.; Lee, B.K.; Jeong, G.S.; Hyun, J.K.; Lee, C.J.; Lee, S.-H. Three-dimensional brain-on-a-chip with an interstitial level of flow and its application as an in vitro model of Alzheimer’s disease. Lab Chip 2015, 15, 141–150. [Google Scholar] [CrossRef]
- Kunze, A.; Giugliano, M.; Valero, A.; Renaud, P. Micropatterning neural cell cultures in 3D with a multi-layered scaffold. Biomaterials 2011, 32, 2088–2098. [Google Scholar] [CrossRef] [PubMed]
- Kilic, O.; Pamies, D.; Lavell, E.; Schiapparelli, P.; Feng, Y.; Hartung, T.; Bal-Price, A.; Hogberg, H.T.; Quinones-Hinojosa, A.; Guerrero-Cazares, H. Brain-on-a-chip model enables analysis of human neuronal differentiation and chemotaxis. Lab Chip 2016, 16, 4152–4162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dauth, S.; Maoz, B.M.; Sheehy, S.P.; Hemphill, M.A.; Murty, T.; Macedonia, M.K.; Greer, A.M.; Budnik, B.; Parker, K.K. Neurons derived from different brain regions are inherently different in vitro: A novel multiregional brain-on-a-chip. J. Neurophysiol. 2016, 117, 1320–1341. [Google Scholar] [CrossRef] [PubMed]
- Mamani, J.B.; Marinho, B.S.; Rego, G.N.d.A.; Nucci, M.P.; Alvieri, F.; Santos, R.S.d.; Ferreira, J.V.M.; Oliveira, F.A.d.; Gamarra, L.F. Magnetic hyperthermia therapy in glioblastoma tumor on-a-Chip model. Einstein 2020, 18. [Google Scholar] [CrossRef]
- Vernetti, L.A.; Senutovitch, N.; Boltz, R.; DeBiasio, R.; Ying Shun, T.; Gough, A.; Taylor, D.L. A human liver microphysiology platform for investigating physiology, drug safety, and disease models. Exp. Biol. Med. 2016, 241, 101–114. [Google Scholar] [CrossRef]
- Prot, J.M.; Aninat, C.; Griscom, L.; Razan, F.; Brochot, C.; Guillouzo, C.G.; Legallais, C.; Corlu, A.; Leclerc, E. Improvement of HepG2/C3a cell functions in a microfluidic biochip. Biotechnol. Bioeng. 2011, 108, 1704–1715. [Google Scholar] [CrossRef]
- Powers, M.J.; Domansky, K.; Kaazempur-Mofrad, M.R.; Kalezi, A.; Capitano, A.; Upadhyaya, A.; Kurzawski, P.; Wack, K.E.; Stolz, D.B.; Kamm, R. A microfabricated array bioreactor for perfused 3D liver culture. Biotechnol. Bioeng. 2002, 78, 257–269. [Google Scholar] [CrossRef]
- Lee, S.-A.; Kang, E.; Ju, J.; Kim, D.-S.; Lee, S.-H. Spheroid-based three-dimensional liver-on-a-chip to investigate hepatocyte–hepatic stellate cell interactions and flow effects. Lab Chip 2013, 13, 3529–3537. [Google Scholar] [CrossRef]
- Lee, P.J.; Hung, P.J.; Lee, L.P. An artificial liver sinusoid with a microfluidic endothelial-like barrier for primary hepatocyte culture. Biotechnol. Bioeng. 2007, 97, 1340–1346. [Google Scholar] [CrossRef]
- Bavli, D.; Prill, S.; Ezra, E.; Levy, G.; Cohen, M.; Vinken, M.; Vanfleteren, J.; Jaeger, M.; Nahmias, Y. Real-time monitoring of metabolic function in liver-on-chip microdevices tracks the dynamics of mitochondrial dysfunction. Proc. Natl. Acad. Sci. USA 2016, 113, E2231–E2240. [Google Scholar] [CrossRef] [Green Version]
- Khetani, S.R.; Bhatia, S.N. Microscale culture of human liver cells for drug development. Nat. Biotechnol. 2008, 26, 120. [Google Scholar] [CrossRef] [PubMed]
- Cho, C.H.; Park, J.; Tilles, A.W.; Berthiaume, F.; Toner, M.; Yarmush, M.L. Layered patterning of hepatocytes in co-culture systems using microfabricated stencils. Biotechniques 2010, 48, 47–52. [Google Scholar] [CrossRef] [PubMed]
- Delalat, B.; Cozzi, C.; Rasi Ghaemi, S.; Polito, G.; Kriel, F.H.; Michl, T.D.; Harding, F.J.; Priest, C.; Barillaro, G.; Voelcker, N.H. Microengineered bioartificial liver chip for drug toxicity screening. Adv. Funct. Mater. 2018, 28, 1801825. [Google Scholar] [CrossRef]
- Kang, Y.B.; Sodunke, T.R.; Lamontagne, J.; Cirillo, J.; Rajiv, C.; Bouchard, M.J.; Noh, M. Liver sinusoid on a chip: Long-term layered co-culture of primary rat hepatocytes and endothelial cells in microfluidic platforms. Biotechnol. Bioeng. 2015, 112, 2571–2582. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Q.; Patel, D.; Kwa, T.; Haque, A.; Matharu, Z.; Stybayeva, G.; Gao, Y.; Diehl, A.M.; Revzin, A. Liver injury-on-a-chip: Microfluidic co-cultures with integrated biosensors for monitoring liver cell signaling during injury. Lab Chip 2015, 15, 4467–4478. [Google Scholar] [CrossRef] [Green Version]
- Kamei, K.-I.; Yoshioka, M.; Terada, S.; Tokunaga, Y.; Chen, Y. Three-dimensional cultured Liver-on-a-Chip with mature hepatocyte-like cells derived from human pluripotent stem cells. Biomed. Microdevices 2019, 21, 73. [Google Scholar] [CrossRef] [Green Version]
- Jang, K.-J.; Mehr, A.P.; Hamilton, G.A.; McPartlin, L.A.; Chung, S.; Suh, K.-Y.; Ingber, D.E. Human kidney proximal tubule-on-a-chip for drug transport and nephrotoxicity assessment. Integr. Biol. 2013, 5, 1119–1129. [Google Scholar] [CrossRef] [PubMed]
- Musah, S.; Mammoto, A.; Ferrante, T.C.; Jeanty, S.S.; Hirano-Kobayashi, M.; Mammoto, T.; Roberts, K.; Chung, S.; Novak, R.; Ingram, M. Mature induced-pluripotent-stem-cell-derived human podocytes reconstitute kidney glomerular-capillary-wall function on a chip. Nat. Biomed. Eng. 2017, 1, 1–12. [Google Scholar] [CrossRef]
- Zhou, M.; Zhang, X.; Wen, X.; Wu, T.; Wang, W.; Yang, M.; Wang, J.; Fang, M.; Lin, B.; Lin, H. Development of a functional glomerulus at the organ level on a chip to mimic hypertensive nephropathy. Sci. Rep. 2016, 6, 31771. [Google Scholar] [CrossRef] [Green Version]
- Wang, J.; Wang, C.; Xu, N.; Liu, Z.-F.; Pang, D.-W.; Zhang, Z.-L. A virus-induced kidney disease model based on organ-on-a-chip: Pathogenesis exploration of virus-related renal dysfunctions. Biomaterials 2019, 219, 119367. [Google Scholar] [CrossRef]
- Wilmer, M.J.; Ng, C.P.; Lanz, H.L.; Vulto, P.; Suter-Dick, L.; Masereeuw, R. Kidney-on-a-chip technology for drug-induced nephrotoxicity screening. Trends Biotechnol. 2016, 34, 156–170. [Google Scholar] [CrossRef] [PubMed]
- Kimura, H.; Yamamoto, T.; Sakai, H.; Sakai, Y.; Fujii, T. An integrated microfluidic system for long-term perfusion culture and on-line monitoring of intestinal tissue models. Lab Chip 2008, 8, 741–746. [Google Scholar] [CrossRef]
- Kim, H.J.; Ingber, D.E. Gut-on-a-Chip microenvironment induces human intestinal cells to undergo villus differentiation. Integr. Biol. 2013, 5, 1130–1140. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, H.J.; Li, H.; Collins, J.J.; Ingber, D.E. Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut-on-a-chip. Proc. Natl. Acad. Sci. USA 2016, 113, E7–E15. [Google Scholar] [CrossRef] [Green Version]
- Shin, W.; Hinojosa, C.D.; Ingber, D.E.; Kim, H.J. Human Intestinal Morphogenesis Controlled by Transepithelial Morphogen Gradient and Flow-Dependent Physical Cues in a Microengineered Gut-on-a-Chip. iScience 2019, 15, 391–406. [Google Scholar] [CrossRef] [Green Version]
- Jahanshahi, M.; Hamdi, D.; Godau, B.; Samiei, E.; Sanchez-Lafuente, C.L.; Neale, K.J.; Hadisi, Z.; Dabiri, S.M.H.; Pagan, E.; Christie, B.R. An Engineered Infected Epidermis Model for In Vitro Study of the Skin’s Pro-Inflammatory Response. Micromachines 2020, 11, 227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohammadi, M.H.; Heidary Araghi, B.; Beydaghi, V.; Geraili, A.; Moradi, F.; Jafari, P.; Janmaleki, M.; Valente, K.P.; Akbari, M.; Sanati-Nezhad, A. Skin diseases modeling using combined tissue engineering and microfluidic technologies. Adv. Healthc. Mater. 2016, 5, 2459–2480. [Google Scholar] [CrossRef] [PubMed]
- Wagner, I.; Materne, E.-M.; Brincker, S.; Süßbier, U.; Frädrich, C.; Busek, M.; Sonntag, F.; Sakharov, D.A.; Trushkin, E.V.; Tonevitsky, A.G. A dynamic multi-organ-chip for long-term cultivation and substance testing proven by 3D human liver and skin tissue co-culture. Lab Chip 2013, 13, 3538–3547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abaci, H.E.; Gledhill, K.; Guo, Z.; Christiano, A.M.; Shuler, M.L. Pumpless microfluidic platform for drug testing on human skin equivalents. Lab Chip 2015, 15, 882–888. [Google Scholar] [CrossRef] [Green Version]
- Alexander, F.A.; Eggert, S.; Wiest, J. Skin-on-a-chip: Transepithelial electrical resistance and extracellular acidification measurements through an automated air-liquid interface. Genes 2018, 9, 114. [Google Scholar] [CrossRef] [Green Version]
- Kwak, B.S.; Jin, S.P.; Kim, S.J.; Kim, E.J.; Chung, J.H.; Sung, J.H. Microfluidic skin chip with vasculature for recapitulating the immune response of the skin tissue. Biotechnol. Bioeng. 2020, 117, 1853–1863. [Google Scholar] [CrossRef] [PubMed]
- Wufuer, M.; Lee, G.; Hur, W.; Jeon, B.; Kim, B.J.; Choi, T.H.; Lee, S. Skin-on-a-chip model simulating inflammation, edema and drug-based treatment. Sci. Rep. 2016, 6, 37471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.; Jin, S.-P.; Kim, Y.K.; Sung, G.Y.; Chung, J.H.; Sung, J.H. Construction of 3D multicellular microfluidic chip for an in vitro skin model. Biomed. Microdevices 2017, 19, 22. [Google Scholar] [CrossRef] [PubMed]
- Jusoh, N.; Ko, J.; Jeon, N.L. Microfluidics-based skin irritation test using in vitro 3D angiogenesis platform. APL Bioeng. 2019, 3, 036101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rezaei Kolahchi, A.; Khadem Mohtaram, N.; Pezeshgi Modarres, H.; Mohammadi, M.H.; Geraili, A.; Jafari, P.; Akbari, M.; Sanati-Nezhad, A. Microfluidic-based multi-organ platforms for drug discovery. Micromachines 2016, 7, 162. [Google Scholar] [CrossRef]
- Abaci, H.E.; Shuler, M.L. Human-on-a-chip design strategies and principles for physiologically based pharmacokinetics/pharmacodynamics modeling. Integr. Biol. 2015, 7, 383–391. [Google Scholar] [CrossRef]
- Shintu, L.; Baudoin, R.G.; Navratil, V.; Prot, J.-M.; Pontoizeau, C.M.; Defernez, M.; Blaise, B.J.; Domange, C.L.; Péry, A.R.; Toulhoat, P. Metabolomics-on-a-chip and predictive systems toxicology in microfluidic bioartificial organs. Anal. Chem. 2012, 84, 1840–1848. [Google Scholar] [CrossRef]
- Vunjak-Novakovic, G.; Bhatia, S.; Chen, C.; Hirschi, K. HeLiVa platform: Integrated heart-liver-vascular systems for drug testing in human health and disease. Stem Cell Res. Ther. 2013, 4, S8. [Google Scholar] [CrossRef] [Green Version]
- Maschmeyer, I.; Hasenberg, T.; Jaenicke, A.; Lindner, M.; Lorenz, A.K.; Zech, J.; Garbe, L.-A.; Sonntag, F.; Hayden, P.; Ayehunie, S. Chip-based human liver–intestine and liver–skin co-cultures–A first step toward systemic repeated dose substance testing in vitro. Eur. J. Pharm. Biopharm. 2015, 95, 77–87. [Google Scholar] [CrossRef] [Green Version]
- Bricks, T.; Paullier, P.; Legendre, A.; Fleury, M.-J.; Zeller, P.; Merlier, F.; Anton, P.M.; Leclerc, E. Development of a new microfluidic platform integrating co-cultures of intestinal and liver cell lines. Toxicol. In Vitro 2014, 28, 885–895. [Google Scholar] [CrossRef]
- Maschmeyer, I.; Lorenz, A.K.; Schimek, K.; Hasenberg, T.; Ramme, A.P.; Hübner, J.; Lindner, M.; Drewell, C.; Bauer, S.; Thomas, A. A four-organ-chip for interconnected long-term co-culture of human intestine, liver, skin and kidney equivalents. Lab Chip 2015, 15, 2688–2699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sung, J.H.; Shuler, M.L. A micro cell culture analog (µCCA) with 3-D hydrogel culture of multiple cell lines to assess metabolism-dependent cytotoxicity of anti-cancer drugs. Lab Chip 2009, 9, 1385–1394. [Google Scholar] [CrossRef] [PubMed]
- Viravaidya, K.; Shuler, M.L. Incorporation of 3T3-L1 cells to mimic bioaccumulation in a microscale cell culture analog device for toxicity studies. Biotechnol. Prog. 2004, 20, 590–597. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.; Zhao, Z.; Rahim, N.A.A.; van Noort, D.; Yu, H. Towards a human-on-chip: Culturing multiple cell types on a chip with compartmentalized microenvironments. Lab Chip 2009, 9, 3185–3192. [Google Scholar] [CrossRef] [PubMed]
- Iori, E.; Vinci, B.; Murphy, E.; Marescotti, M.C.; Avogaro, A.; Ahluwalia, A. Glucose and fatty acid metabolism in a 3 tissue in-vitro model challenged with normo-and hyperglycaemia. PLoS ONE 2012, 7, e34704. [Google Scholar] [CrossRef] [Green Version]
- Ucciferri, N.; Sbrana, T.; Ahluwalia, A. Allometric scaling and cell ratios in multi-organ in vitro models of human metabolism. Front. Bioeng. Biotechnol. 2014, 2, 74. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.S.; Aleman, J.; Shin, S.R.; Kilic, T.; Kim, D.; Shaegh, S.A.M.; Massa, S.; Riahi, R.; Chae, S.; Hu, N. 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. [Google Scholar] [CrossRef] [Green Version]
- Roberts, D.A.; Kantarjian, H.M.; Steensma, D.P. Contract research organizations in oncology clinical research: Challenges and opportunities. Cancer 2016, 122, 1476–1482. [Google Scholar] [CrossRef] [Green Version]
- Valente, A. MEMS Devices in Agriculture. In Advanced Mechatronics and MEMS Devices II; Springer: Berlin/Heidelberg, Germany, 2017; pp. 367–385. [Google Scholar]
- Kim, Y.; Abafogi, A.T.; Tran, B.M.; Kim, J.; Lee, J.; Chen, Z.; Bae, P.K.; Park, K.; Shin, Y.-B.; van Noort, D. Integrated Microfluidic Preconcentration and Nucleic Amplification System for Detection of Influenza A Virus H1N1 in Saliva. Micromachines 2020, 11, 203. [Google Scholar] [CrossRef] [Green Version]
- Li, L.; Miao, B.; Li, Z.; Sun, Z.; Peng, N. Sample-to-Answer Hepatitis B Virus DNA Detection from Whole Blood on a Centrifugal Microfluidic Platform with Double Rotation Axes. ACS Sens. 2019, 4, 2738–2745. [Google Scholar] [CrossRef]
- Mejía-Salazar, J.R.; Rodrigues Cruz, K.; Materón Vásques, E.M. Microfluidic Point-of-Care Devices: New Trends and Future Prospects for eHealth Diagnostics. Sensors 2020, 20, 1951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, J.; Pang, J.; Chen, Y.; Dong, Q.; Sheng, J.; Luo, Y.; Lu, Y.; Lin, B.; Liu, T. Application of microfluidic chips in separation and analysis of extracellular vesicles in liquid biopsy for cancer. Micromachines 2019, 10, 390. [Google Scholar] [CrossRef] [Green Version]
- Sharma, S.; Kumar, V.; Chawla, A.; Logani, A. Rapid detection of SARS-CoV-2 in saliva: Can an endodontist take the lead in point-of-care COVID-19 testing? Int. Endod. J. 2020, 53, 1017–1019. [Google Scholar] [CrossRef]
- Yang, T.; Wang, Y.-C.; Shen, C.-F.; Cheng, C.-M. Point-of-Care RNA-Based Diagnostic Device for COVID-19; Multidisciplinary Digital Publishing Institute: Basel, Switzerland, 2020. [Google Scholar]
- Udugama, B.; Kadhiresan, P.; Kozlowski, H.N.; Malekjahani, A.; Osborne, M.; Li, V.Y.; Chen, H.; Mubareka, S.; Gubbay, J.B.; Chan, W.C. Diagnosing COVID-19: The disease and tools for detection. ACS Nano 2020, 14, 3822–3835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Riahi, R.; Tamayol, A.; Shaegh, S.A.M.; Ghaemmaghami, A.M.; Dokmeci, M.R.; Khademhosseini, A. Microfluidics for advanced drug delivery systems. Curr. Opin. Chem. Eng. 2015, 7, 101–112. [Google Scholar] [CrossRef] [PubMed]
- Patra, B.; Lafontaine, J.; Bavoux, M.; Zerouali, K.; Glory, A.; Ahanj, M.; Carrier, J.-F.; Gervais, T.; Wong, P. On-chip combined radiotherapy and chemotherapy testing on soft-tissue sarcoma spheroids to study cell death using flow cytometry and clonogenic assay. Sci. Rep. 2019, 9, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Mokhtari, R.B.; Homayouni, T.S.; Baluch, N.; Morgatskaya, E.; Kumar, S.; Das, B.; Yeger, H. Combination therapy in combating cancer. Oncotarget 2017, 8, 38022. [Google Scholar] [CrossRef] [Green Version]
- Zhao, M.; Wei, D.-Q. Rare diseases: Drug discovery and informatics resource. Interdiscip. Sci. Comput. Life Sci. 2018, 10, 195–204. [Google Scholar] [CrossRef]
- De Mello, C.P.P.; Rumsey, J.; Slaughter, V.; Hickman, J.J. A human-on-a-chip approach to tackling rare diseases. Drug Discov. Today 2019, 24, 2139–2151. [Google Scholar] [CrossRef]
Company Name | Materials and Manufacturing | Analytes | Applications | Sample Types | Signal Detection | Highlights of Technology Suited for POC |
---|---|---|---|---|---|---|
Abaxis | Plastic disc | Small molecules, proteins | Blood chemistries (e.g., metaboliteselectrolytes) | Whole blood | Absorbance | Compact analyzer, injection-molded plastic discs, no pre-processing of sample |
Advanced Liquid Logic | Glass, insulated electrodes | Small molecules, proteins, nucleic acids | HIV/AIDS, Iysosomal storage disease | Whole blood | Fluorescence, chemiluminescence | Sample pre-processing compact, benchtop analyzer, manipulation of nano-and micro-droplets |
Alere(formerly inverness Medical) | Plastic and elastically deformable materials | Proteins, cells | HIV/AIDS, clotting time | Whole blood | Fluorescence | Disposable cartridge, portable analyzer, automated image-based immune hematology test |
Biosite (Alere) | Strip with textured microstructures | Small molecules, proteins | Cardiovascular disease, drugs of abuse, waterborne parasites | Whole blood, plasma | Fluorescence | Portable reader, disposable capillary-driven microfluidic test strips |
Cepheid | Disposable plastic cartridge | Nucleic acids | Respiratory infections (bacterial and viral), cancer | Whole blood, sputum | Fluorescence (with molecular beacons) | Disposable cards with benchtop analyzer, on-card sample processing (sputum) |
Daktari Diagnostics | Plastic cartridge | Cells | HIV/AIDS | Whole blood | Electrochemical (impedance spectroscopy) | Handheld instrument, label-free electrochemical sensing of captured cell lysate |
Diagnostic For All | Paper | Small molecules, Proteins | Liver damage from HIV/AIDS medication | Whole blood, urine | Colorimetric | Instrument-free tests based on paper, capillary-driven microfluidics, colorimetric readout |
Epocal (Alere) | Film, epoxy laminates | Small molecules | Blood chemistries | Whole blood | Electrochemical, chemiluminescence | Self-contained cards, patterned electrodes for sensing, wireless data transmission |
Focus Dx (Quest) | Plastic (polypropylene) | Nucleic acids | Flu, intestinal pathogens | Nasal and pharyngeal swabs | Fluorescence | Portable detector, discs with on-board extraction |
HandyLab (BD) | Disposable cartridges | Nucleic acids | Bacterial infections and drug susceptibility testing | Vaginal, rectal, nasal swabs | Fluorescence (with molecular beacons) | Disposable cards with integrated heating, detection, sample processing in a portable instrument |
i-STAT Corp (Abbott) | Plastic cartridge with silicon microchip | Small molecules | Blood chemistries, coagulation, cardiac markers | Whole blood, urine | Electrochemical (potentiometry, amperometry, conductivity) | Portable analyzer, capillary-driven microfluidics, thin-film electrodes for detection |
Micronics (Sony) | Plastic, laminates, paper | Proteins, nucleic acids | Malaria, shiga toxin-producing E-coli, ABO blood typing | Whole blood, stool | Absorbance, colorimetry | Disposable cartridges composed of thin-film laminates and injection-molding |
Philips | Plastic cartridge | Nucleic acids, small molecules, proteins | Cardiac damage, drugs of abuse, hormones | Whole blood, saliva | Optical (frustrated total internal reflectance) | Handheld reader with self-concentration of magnetic nanoparticles for rapid analysis |
TearLab | Polycarbonate | Small molecules | Dry eye disease (tear osmolarity), ocular allergy (IgE antibodies) | Tear | Electrochemical | Portable osmolarity reader with disposable cards; capillary-driven flow, gold electrodes for detection, results in 5 s |
Cell Culture Methods | Advantages | Disadvantages |
---|---|---|
2D cell culture |
|
|
3D cell culture models |
|
|
Microfluidic cell culture platforms |
|
|
Company’s Name | Summary of Specialties | Applications | Cell Source | Highlight of the Technology | Year |
---|---|---|---|---|---|
AlveoliX | OOC, Lung-on-a-chip | Drug discovery, disease modeling | Human cell lines | In vitro models inspired by nature, reproduce lung breathing motion, elastic and ultrathin membrane | 2015 |
AxoSim | OOC, nerve-on-a-chip | Preclinical testing, 3D cell culture, neurotoxicity tests, neurodegenerative diseases | Primary cultures, Organoids | Biomimetic human tissues, combination of neurons, astrocytes, and oligodendrocytes. | 2014 |
BEOnChip | OOC | Disease modeling, in vitro tests, drug screening | Human cells | Long-term 2D or 3D culture under flow condition, 2D-3D co-culture, simulation of physiological environments involving flow and shear stress | 2016 |
BiomimX | OOC, heart-on-a-chip, Cartilage-on-a-chip | Drug screening, drug cardiotoxicity, anti-arrhythmic drug efficiency, discovery anti- osteoarthritic drugs | cardiomyocytes derived from human iPSCs, human cells | 3D co-culture, mechanical stimulations, human cardiac tissue, human osteoarthritic cartilage, customized OOC | 2017 |
BI/OND | OOC, BI/OND’s microfluidic plate | In vitro tests, Drug discovery | Human cells, Organoids, patient derived cells or tissues | Dynamic cell culture environment by providing mechanical stimulation and continuous fluid flow, two compartments connected by a porous membrane BI/OND’s plate to run up to six cultures in parallel, 3D and 2D models | 2017 |
CN Bio Innovations | OOC, ), liver-on-a-chip, Body-on-a-chip (7-OOC ) | Human physiology modelling, liver diseases modelling, Preclinical drug discovery, toxicity tests, drug metabolism | Primary human cells, Tissue or Organ Slices, IPSCs, Immortalised cell lines | Multi organ studies, portable and compact device, programmable flow rate, open well plates | 2009 |
Emulate, Inc. | OOC, Lung, Bone marrow, kidney, brain, blood vessels and intestine-on-a-chip | Personalized medicine, disease modelling, drug screening, study human physiology | - | Organ-Chips personalized with individual patients’ stem cells, stretchable biochip, flexible and dynamic environment by fluid flow and mechanical stretch | 2014 |
Hesperos | OOC, multi-organ-on-a-chip ( heart, liver, lung, brain, skin, muscle, kidney, pancreas, bone marrow) | In vitro tests, drug discovery, toxicity tests, pharmacokinetic/ pharmacodynamic modeling | human stem cells | Pumpless platform, recreate muscle and tissue function, neural and inter-organ communication, customized human-on-a-chip platform, possibility to add immune cells in multi-organ-platform | 2015 |
MIMETAS | OOC | Disease modelling, drug testing, toxicity tests, personalized medicine | Human cells, patient derived cells or tissues | OrganoPlates (a microfluidic 3D cell culture plate), 3D co-culture, biomimetic, compatible, easy to use | 2013 |
Nortis | OOC, kidney, brain, heart, liver, immune system and blood vessels-on-a-chip | Disease modeling, cancer study, drug testing, study Alzheimer’s disease and ageing, toxicity tests | Human derived tissue models | Perfusion system, standard cell culture incubator, | 2012 |
SynVivo, Inc | OOC, blood-brain-barrier-on-a-chip | Drug discovery, toxicity test, targeted drug delivery, cancer researches | Human cells | Mimic microvascular environment, dynamic environment, real-time visualization, controlled condition, 3D co-culture model | 2014 |
TARA Biosystems | OOC, heart-on-a-chip | Cardiac Toxicology, Precision Cardiology, Heart Failure Drug Discovery, Drug development, study healthy and disease models | iPSCs derived cardiomyocytes | Cardiac tissue models, patient derived disease models | 2014 |
TissUse | OOC, body-on-a-chip | Toxicity tests, disease modeling, personalized medicine, drug development, application in pharmaceutical and cosmetic researches | Cell lines, human primary cells, biopsies | Multi-organ platforms, rapid prototyping, compatible with tissue imaging, application of physiological sheer stress, long-term performance | 2010 |
Technology | LOC | OOC |
---|---|---|
Applications in life science |
|
|
Advantages |
|
|
Drawbacks |
|
|
Size |
|
|
Materials |
|
|
Biological samples |
|
|
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Azizipour, N.; Avazpour, R.; Rosenzweig, D.H.; Sawan, M.; Ajji, A. Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. Micromachines 2020, 11, 599. https://doi.org/10.3390/mi11060599
Azizipour N, Avazpour R, Rosenzweig DH, Sawan M, Ajji A. Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. Micromachines. 2020; 11(6):599. https://doi.org/10.3390/mi11060599
Chicago/Turabian StyleAzizipour, Neda, Rahi Avazpour, Derek H. Rosenzweig, Mohamad Sawan, and Abdellah Ajji. 2020. "Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip" Micromachines 11, no. 6: 599. https://doi.org/10.3390/mi11060599
APA StyleAzizipour, N., Avazpour, R., Rosenzweig, D. H., Sawan, M., & Ajji, A. (2020). Evolution of Biochip Technology: A Review from Lab-on-a-Chip to Organ-on-a-Chip. Micromachines, 11(6), 599. https://doi.org/10.3390/mi11060599