AccScience Publishing / IJB / Volume 10 / Issue 2 / DOI: 10.36922/ijb.1851
Cite this article
21
Download
448
Views
Journal Browser
Volume | Year
Issue
Search
News and Announcements
View All
RESEARCH ARTICLE

Bioprinted autologous human skin equivalents for in vitro testing of therapeutic antibodies

Mahid Ahmed1,2 David Hill3,4 Shaheda Ahmed2 Stefan Przyborski5,6 Kenneth Dalgarno1* Anne Dickinson2,4
Show Less
1 School of Engineering, Newcastle University, Newcastle upon Tyne, NE3 1PS, United Kingdom
2 Alcyomics Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne, NE4 5BX, United Kingdom
3 Faculty of Health Sciences and Wellbeing, Sunderland University, Sunderland, SR1 3SD, United Kingdom
4 Translational & Clinical Research Institute, Faculty of Medical Science, Medical School, Newcastle University, Newcastle upon Tyne, NE2 4HH, United Kingdom
5 Department of Bioscience, Durham University, South Road, Durham, DH1 3LE, United Kingdom
6 Reprocell Europe Ltd., NETPark Plexus, Thomas Wright Way, Sedgefield, Co. Durham, TS21 3FD, United Kingdom
IJB 2024, 10(2), 1851 https://doi.org/10.36922/ijb.1851
Submitted: 17 September 2023 | Accepted: 10 January 2024 | Published: 11 March 2024
© 2024 by the Author(s). This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

In recent years, advances in tissue engineering have brought forward the accessibility of human skin equivalents for in vitro applications; however, the availability of human-based engineered tissue models suitable for high-throughput screening of biologics remains limited. Here, we report a method of manufacturing fully autologous (with both fibroblasts and keratinocytes from the same donor) human skin equivalents for determining preclinical therapeutic antibody adverse immune reactions in vitro. Using a combination of precise solenoid microvalve-based bioprinting and 96-well scale Alvetex inserts, autologous skin cells were bioprinted and cultured to develop a scalable approach to manufacturing skin equivalents. We demonstrated that fibroblasts and keratinocytes can be bioprinted with a high degree of precision while maintaining viability post printing. Histological staining showed that the bioprinted 96-well based skin equivalents were comparable to human skin. The fully autologous human skin equivalents were co-cultured in vitro with autologous peripheral blood monocytes with and without muromonab-CD3 (OKT3) and natalizumab (Tysabri), biologics which are known to cause and inhibit adverse immune reactions (type IV hypersensitivity), respectively. Analysis of supernatants from skin-equivalent monocyte co-cultures revealed significant proinflammatory cytokine responses (such as interferon gamma) in co-cultures treated with OKT3 when compared to Tysabri and negative controls. Consequently, this study provides proof of concept that through a combination of bioprinting and Alvetex scaffold-based culture systems, scalable human skin equivalents can be manufactured for high-throughput identification of adverse immune reactions during preclinical stages of the drug development process.

Keywords
Autologous tissue models
Skin-equivalent models
Microvalve bioprinting
Transwell culture
Funding
This research was supported by the EPSRC Centre for Doctoral Training in Additive Manufacturing and 3D Printing (EP/L01534X/1), and by Innovate UK through Project 103597 (Novel manufacture and commercialization of a 96-well 3D skin model for drug and toxicology testing).
References
  1. Stebbings R, Findlay L, Edwards C, et al. “Cytokine storm” in the phase I trial of monoclonal antibody TGN1412: better understanding the causes to improve preclinical testing of immunotherapeutics. J Immunol. 2007;179(5):3325-3331. doi: 10.4049/jimmunol.179.5.3325
  2. Hay M, Thomas D, Craighead J, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32:40-51. doi: 10.1038/nbt.2786
  3. Pound P, Ritskes-Hoitinga M. Is it possible to overcome issues of external validity in preclinical animal research? Why most animal models are bound to fail. J Transl Med. 2018;16:304. doi: 10.1186/s12967-018-1678-1
  4. Panoskaltsis N, McCarthy NE, Knight SC. Myelopoiesis of acute inflammation: lessons from TGN1412-induced cytokine storm. Cancer Immunol Immunother. 2021;70:1155-1160. doi: 10.1007/s00262-020-02702-9
  5. DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. J Health Econ. 2016;47:20-33. doi: 10.1016/j.jhealeco.2016.01.012
  6. Mahlich J, Bartol A, Dheban S. Can adaptive clinical trials help to solve the productivity crisis of the pharmaceutical industry? - a scenario analysis. Health Econ Rev. 2021;11:4. doi: 10.1186/s13561-021-00302-6
  7. Hughes JP, Rees S, Kalindjian SB, Philpott KL. Principles of early drug discovery. Br J Pharmacol. 2011;162(6):1239-1249. doi: 10.1111/j.1476-5381.2010.01127.x
  8. Ahmed SS, Whritenour J, Ahmed MM, et al. Evaluation of a human in vitro skin test for predicting drug hypersensitivity reactions. Toxicol Appl Pharmacol. 2019;369:39-48. doi: 10.1016/j.taap.2019.02.005
  9. Lawrence E, Sims J, Gander A, et al. The barriers and motivators to using human tissues for research: the views of UK-based biomedical researchers. Biopreserv Biobank. 2020;18(4):266-273. doi: 10.1089/bio.2019.0138
  10. Harley WS, Li CC, Toombs J, et al. Advances in biofabrication techniques towards functional bioprinted heterogeneous engineered tissues: a comprehensive review. Bioprinting. 2021;23:e00147. doi: 10.1016/j.bprint.2021.e00147
  11. di Marzio N, Eglin D, Serra T, Moroni L. Bio-Fabrication: convergence of 3D bioprinting and nano-biomaterials in tissue engineering and regenerative medicine. Front Bioeng Biotechnol. 2020;8. doi: 10.3389/fbioe.2020.00326
  12. Groll J, Boland T, Blunk T, et al. Biofabrication: reappraising the definition of an evolving field. Biofabrication. 2016;8(1):013001. doi: 10.1088/1758-5090/8/1/013001
  13. Koch L, Deiwick A, Franke A, et al. Laser bioprinting of human induced pluripotent stem cells-the effect of printing and biomaterials on cell survival, pluripotency, and differentiation. Biofabrication. 2018;10(3):035005. doi: 10.1088/1758-5090/aab981
  14. Cubo N, Garcia M, Del Cañizo JF, Velasco D, Jorcano JL. 3D bioprinting of functional human skin: production and in vivo analysis. Biofabrication. 2016;9(1):015006. doi: 10.1088/1758-5090/9/1/015006
  15. Murphy S, Atala A. 3D bioprinting of tissues and organs. Nat Biotechnol. 2014;32:773-785. doi: 10.1038/nbt.2958
  16. Solis LH, Ayala Y, Portillo S, Varela-Ramirez A, Aguilera R, Boland T. Thermal inkjet bioprinting triggers the activation of the VEGF pathway in human microvascular endothelial cells in vitro. Biofabrication. 2019;11(4):045005. doi: 10.1088/1758-5090/ab25f9
  17. Dudman JPR, Ferreira AM, Gentile P, et al. Reliable inkjet printing of chondrocytes and MSCs using reservoir agitation. Biofabrication. 2020;12(4):045024. doi: 10.1088/1758-5090/aba2f8 
  18. Li X, Liu B, Pei B, et al. Inkjet bioprinting of biomaterials. Chem Rev. 2020;120(19):10793-10833. doi: 10.1021/acs.chemrev.0c00008
  19. Faulkner-Jones A, Fyfe C, Cornelissen DJ, et al. Bioprinting of human pluripotent stem cells and their directed differentiation into hepatocyte-like cells for the generation of mini-livers in 3D. Biofabrication. 2015;7(4):044102. doi: 10.1088/1758-5090/7/4/044102
  20. Dudman J, Ferreira AM, Gentile P, Wang X, Dalgarno K. Microvalve bioprinting of MSC-chondrocyte co-cultures. Cells. 2021;10(12):3329. doi: 10.3390/cells10123329
  21. Ng WL, Qi JTZ, Yeong WY, Naing MW. Proof-of-concept: 3D bioprinting of pigmented human skin constructs. Biofabrication. 2018;10(2):025005. doi: 10.1088/1758-5090/aa9e1e
  22. Yang X, Lu Z, Wu H, Li W, Zheng L, Zhao J. Collagen-alginate as bioink for three-dimensional (3D) cell printing based cartilage tissue engineering. Mater Sci Eng C. 2018;83:195-201. doi: 10.1016/j.msec.2017.09.002
  23. Fedorovich NE, Kuipers E, Gawlitta D, Dhert WJA, Alblas J. Scaffold porosity and oxygenation of printed hydrogel constructs affect functionality of embedded osteogenic progenitors. Tissue Eng Part A. 2011;17(19-20):2473-2486. doi: 10.1089/ten.TEA.2011.0001
  24. Alonzo M, AnilKumar S, Roman B, Tasnim N, Joddar B. 3D bioprinting of cardiac tissue and cardiac stem cell therapy. Transl Res. 2019;211:64-83. doi: 10.1016/j.trsl.2019.04.004
  25. Albanna M, Binder KW, Murphy SV, et al. In situ bioprinting of autologous skin cells accelerates wound healing of extensive excisional full-thickness wounds. Sci Rep. 2019;9(1):1856. doi: 10.1038/s41598-018-38366-w
  26. Zhou Y, Qin R, Chen T, Zhang K, Gui J. 3D bioprinting modified autologous matrix-induced chondrogenesis (AMIC) technique for repair of cartilage defects. Mater Des. 2021;203. doi: 10.1016/j.matdes.2021.109621
  27. Hill DS, Robinson ND, Caley MP, et al. A novel fully humanized 3D skin equivalent to model early melanoma invasion. Mol Cancer Ther. 2015;14(11):2665-2673. doi: 10.1158/1535-7163.MCT-15-0394
  28. Choudhury S, Das A. Advances in generation of three-dimensional skin equivalents: pre-clinical studies to clinical therapies. Cytotherapy. 2021;23(1):P1-9. doi: 10.1016/j.jcyt.2020.10.001
  29. Berger A. Th1 and Th2 responses: what are they? BMJ. 2000;321:424. doi: 10.1136/bmj.321.7258.424
  30. Swain SL, Weinberg AD, English M, Huston G. IL-4 directs the development of Th2-like helper effectors. J Immunol. 1990;145(11):3796-3806.
  31. Gaston RS, Deierhoi MH, Patterson T, et al. OKT3 first-dose reaction: association with T cell subsets and cytokine release. Kidney Int. 1991;39(1):141-148. doi: 10.1038/ki.1991.18
  32. Römer PS, Berr S, Avota E, et al. Preculture of PBMCs at high cell density increases sensitivity of T-cell responses, revealing cytokine release by CD28 superagonist TGN1412. Blood. 2011;118(26):6772-6782. doi: 10.1182/blood-2010-12-319780
  33. Yoshizaki, K, Nishimoto N, Matsumoto K, et al. Interleukin 6 and expression of its receptor on epidermal keratinocytes. Cytokine. 1990;2(5):381-387. doi: 10.1016/1043-4666(90)90069-6
  34. Mazzocchi A, Soker S, Skardal A. 3D bioprinting for high-throughput screening: drug screening, disease modeling, and precision medicine applications. Appl Phys Rev. 2019;6(1):011302. doi: 10.1063/1.5056188
  35. Mandrycky C, Wang Z, Kim K, Kim DH. 3D bioprinting for engineering complex tissues. Biotechnol Adv. 2016;34(4): 422-434. doi: 10.1016/j.biotechadv.2015.12.011 
  36. Lee V, Singh G, Trasatti JP, et al. Design and fabrication of human skin by three-dimensional bioprinting. Tissue Eng Part C. 2014;20(6):473-484. doi: 10.1089/ten.TEC.2013.0335
  37. Michael S, Sorg H, Peck CT, et al. Tissue engineered skin substitutes created by laser-assisted bioprinting form skin-like structures in the dorsal skin fold chamber in mice. PLoS One. 2013;8(3):e57741. doi: 10.1371/journal.pone.0057741
  38. Weng T, Zhang W, Xia Y, et al. 3D bioprinting for skin tissue engineering: current status and perspectives. J Tissue Eng. 2021;12:20417314211028574. doi: 10.1177/20417314211028574
Conflict of interest
The authors declare no conflicts of interest.
Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing