AccScience Publishing / IJB / Volume 9 / Issue 4 / DOI: 10.18063/ijb.723
RESEARCH ARTICLE

Using 3D-bioprinted models to study pediatric neural crest-derived tumors

Colin H. Quinn1† Andee M. Beierle2† Janet R. Julson1 Michael E. Erwin1 Hasan Alrefai2 Hooper R. Markert1 Jerry E. Stewart1 Sara Claire Hutchins3 Laura V. Bownes1 Jamie M. Aye3 Elizabeth Mroczek-Musulman4 Patricia H. Hicks4 Karina J. Yoon5 Christopher D. Willey2* Elizabeth A. Beierle1*
Show Less
1 Division of Pediatric Surgery, Department of Surgery, University of Alabama, Birmingham, Birmingham, AL, 35205, USA
2 Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL, 35205, USA
3 Division of Pediatric Hematology Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
4 Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
5 Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
(This article belongs to the Special Issue Bioprinting process for tumor model development)
© Invalid date 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

The use of three-dimensional (3D) bioprinting has remained at the forefront of tissue engineering and has recently been employed for generating bioprinted solid tumors to be used as cancer models to test therapeutics. In pediatrics, neural crest-derived tumors are the most common type of extracranial solid tumors. There are only a few tumor-specific therapies that directly target these tumors, and the lack of new therapies remains detrimental to improving the outcomes for these patients. The absence of more efficacious therapies for pediatric solid tumors, in general, may be due to the inability of the currently employed preclinical models to recapitulate the solid tumor phenotype. In this study, we utilized 3D bioprinting to generate neural crest-derived solid tumors. The bioprinted tumors consisted of cells from established cell lines and patient-derived xenograft tumors mixed with a 6% gelatin/1% sodium alginate bioink. The viability and morphology of the bioprints were analyzed via bioluminescence and immunohisto chemistry, respectively. We compared the bioprints to traditional twodimensional (2D) cell culture under conditions such as hypoxia and therapeutics. We successfully produced viable neural crest-derived tumors that retained the histology and immunostaining characteristics of the original parent tumors. The bioprinted tumors propagated in culture and grew in orthotopic murine models. Furthermore, compared to cells grown in traditional 2D culture, the bioprinted tumors were resistant to hypoxia and chemotherapeutics, suggesting that the bioprints exhibited a phenotype that is consistent with that seen clinically in solid tumors, thus potentially making this model superior to traditional 2D culture for preclinical investigations. Future applications of this technology entail the potential to rapidly print pediatric solid tumors for use in highthroughput drug studies, expediting the identification of novel, individualized therapies.

Keywords
3D bioprinting
Neuroblastoma
Neuroendocrine; Pediatrics
Targeted therapy
Patient-derived xenografts
References

1. Siegel RL, Miller KD, Fuchs HE, et al., 2022, Cancer statistics. CA Cancer J Clin, 72(1):7–33. https://doi.org/10.3322/caac.21708 

2. Colon NC, Chung DH, 2011, Neuroblastoma. Adv Pediatr, 58(1):297–311. https://doi.org/10.1016/j.yapd.2011.03.011 

3. Navalkele P, O’Dorisio MS, O’Dorisio TM, et al., 2011, Incidence, survival, and prevalence of neuroendocrine tumors versus neuroblastoma in children and young adults: Nine standard SEER registries, 1975-2006. Pediatr Blood Cancer, 56(1):50–57. https://doi.org/10.1002/pbc.22559 

4. LeSavage BL, Suhar RA, Broguiere N, et al., 2022, Next-generation cancer organoids. Nat Mater, 21(2):143–159. https://doi.org/10.1038/s41563-021-01057-5 

5. Kang Y, Datta P, Shanmughapriya S, et al., 2020, 3D bioprinting of tumor models for cancer research. ACS Appl Bio Mater, 3(9):5552–5573. https://doi.org/10.1021/acsabm.0c00791 

6. Zhuang P, Chiang YH, Fernanda MS, et al., 2021, Using spheroids as building blocks towards 3D bioprinting of tumor microenvironment. Int J Bioprint, 7(4):444. https://doi.org/10.18063/ijb.v7i4.444 

7. Neufeld L, Yeini E, Reisman N, et al., 2021, Microengineered perfusable 3D-bioprinted glioblastoma model for in vivo mimicry of tumor microenvironment. Sci Adv, 7(34):eabi9119. https://doi.org/10.1126/sciadv.abi9119
 
8. Langer EM, Allen-Petersen BL, King SM, et al., 2019, Modeling tumor phenotypes in vitro with three-dimensional bioprinting. Cell Rep, 26(3): 608–623 e6. https://doi.org/10.1016/j.celrep.2018.12.090 

9. Stafman LL, Williams AP, Marayati R, et al., 2019, Focal adhesion kinase inhibition contributes to tumor cell survival and motility in neuroblastoma patient-derived xenografts. Sci Rep, 9(1):13259. https://doi.org/10.1038/s41598-019-49853-z 

10. Quinn CH, Beierle AM, Williams AP, et al., 2021, Downregulation of PDGFRss signaling overcomes crizotinib resistance in a TYRO3 and ALK mutated neuroendocrine-like tumor. Transl Oncol, 14(7):101099. https://doi.org/10.1016/j.tranon.2021.101099 

11. Marayati R, Bownes LV, Quinn CH, et al., 2021, Novel second-generation rexinoid induces growth arrest and reduces cancer cell stemness in human neuroblastoma patient-derived xenografts. J Pediatr Surg, 56(6):1165–1173. https://doi.org/10.1016/j.jpedsurg.2021.02.041 

12. Stafman LL, Mruthyunjayappa S, Waters AM, et al., 2018, Targeting PIM kinase as a therapeutic strategy in human hepatoblastoma. Oncotarget, 9(32):22665–22679. https://doi.org/10.18632/oncotarget.25205 

13. Quinn CH, Beierle AM, Hutchins SC, et al., 2022, Targeting high-risk neuroblastoma patient-derived xenografts with oncolytic virotherapy. Cancers (Basel), 14(3):762. https://doi.org/10.3390/cancers14030762

14. Tomayko MM, Reynolds CP, 1989, Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol, 24(3):148–154. https://doi.org/10.1007/BF00300234 

15. Hirabayashi K, Zamboni G, Nishi T, et al., 2013 Histopathology of gastrointestinal neuroendocrine neoplasms. Front Oncol, 3:2. https://doi.org/10.3389/fonc.2013.00002 

16. Bownes LV, Williams AP, Marayati R, et al., 2021, Serine-threonine kinase receptor-associated protein (STRAP) knockout decreases the malignant phenotype in neuroblastoma cell lines. Cancers (Basel), 13(13):3201. https://doi.org/10.3390/cancers13133201 

17. Hockel M,Vaupel P, 2001, Biological consequences of tumor hypoxia. Semin Oncol, 28(2 Suppl 8):36–41. https://www.ncbi.nlm.nih.gov/pubmed/11395851. 

18. Jogi A, Ora I, Nilsson H, et al., 2002, Hypoxia alters gene expression in human neuroblastoma cells toward an immature and neural crest-like phenotype. Proc Natl Acad Sci U S A, 99(10):7021–7026. https://doi.org/10.1073/pnas.102660199 

19. Braekeveldt N, Bexell D, 2018, Patient-derived xenografts as preclinical neuroblastoma models. Cell Tissue Res, 372(2):233–243. https://doi.org/10.1007/s00441-017-2687-8 

20. Neel DV, Shulman DS, DuBois SG, 2019, Timing of first-in-child trials of FDA-approved oncology drugs. Eur J Cancer, 112:49–56. https://doi.org/10.1016/j.ejca.2019.02.011 

21. Byron SA, Hendricks WPD, Nagulapally AB, et al., 2021, Genomic and transcriptomic analysis of relapsed and refractory childhood solid tumors reveals a diverse molecular landscape and mechanisms of immune evasion. Cancer Res, 81(23):5818–5832. https://doi.org/10.1158/0008-5472.CAN-21-1033 

22. Aguirre AJ, Nowak JA, Camarda ND, et al., 2018, Real-time genomic characterization of advanced pancreatic cancer to enable precision medicine.Cancer Discov, 8(9):1096–1111. https://doi.org/10.1158/2159-8290.CD-18-0275 

23. Reda M, Richard C, Bertaut A, et al., 2020, Implementation and use of whole exome sequencing for metastatic solid cancer. EBioMedicine, 51:102624. https://doi.org/10.1016/j.ebiom.2019.102624 

24. Murphy JM, Lim IIP, Farber BA, et al., 2016, Salvage rates after progression of high-risk neuroblastoma with a soft tissue mass. J Pediatr Surg, 51(2):285–288. https://doi.org/10.1016/j.jpedsurg.2015.10.075 

25. Cobain EF, Wu YM, Vats P, et al., 2021, Assessment of clinical benefit of integrative genomic profiling in advanced solid tumors.JAMA Oncol, 7(4):525–533. https://doi.org/10.1001/jamaoncol.2020.7987 

26. Kapalczynska M, Kolenda T, Przybyla W, et al., 2018, 2D and 3D cell cultures—A comparison of different types of cancer cell cultures. Arch Med Sci, 14(4):910–919. https://doi.org/10.5114/aoms.2016.63743 

27. Stylianopoulos T, Jain RK, 2013, Combining two strategies to improve perfusion and drug delivery in solid tumors. Proc Natl Acad Sci U S A, 110(46):18632–18637. https://doi.org/10.1073/pnas.1318415110 

28. Brown JM, Wilson WR, 2004, Exploiting tumour hypoxia in cancer treatment. Nat Rev Cancer, 4(6):437–447. https://doi.org/10.1038/nrc1367 

29. Bhandari V, Hoey C, Liu LY, et al., 2019, Molecular landmarks of tumor hypoxia across cancer types. Nat Genet, 51(2):308–318. https://doi.org/10.1038/s41588-018-0318-2 

30. Li Y, Li B, Li W, et al., 2020, Murine models of IDH-wild-type glioblastoma exhibit spatial segregation of tumor initiation and manifestation during evolution. Nat Commun, 11(1):3669. https://doi.org/10.1038/s41467-020-17382-3
 
31. Braekeveldt N, von Stedingk K, Fransson S, et al., 2018, Patient-derived xenograft models reveal intratumor heterogeneity and temporal stability in neuroblastoma. Cancer Res, 78(20):5958–5969. https://doi.org/10.1158/0008-5472.CAN-18-0527 

32. Dong R, Yang R, Zhan Y, et al., 2020, Single-cell characterization of malignant phenotypes and developmental trajectories of adrenal neuroblastoma. Cancer Cell, 38(5):716–733.e6. https://doi.org/10.1016/j.ccell.2020.08.014 

33. Manas A, Aaltonen K, Andersson N, et al., 2022, Clinically relevant treatment of PDX models reveals patterns of neuroblastoma chemoresistance.Sci Adv, 8(43):eabq4617. https://doi.org/10.1126/sciadv.abq4617 

34. Pellegrini E, Desando G, Petretta M, et al., 2022, A 3D collagen-based bioprinted model to study osteosarcoma invasiveness and drug response. Polymers (Basel), 14(19):4070. https://doi.org/10.3390/polym14194070 

35. Tang M, Xie Q, Gimple RC, et al., 2020, Three-dimensional bioprinted glioblastoma microenvironments model cellular dependencies and immune interactions. Cell Res, 30(10):833–853. https://doi.org/10.1038/s41422-020-0338-1

36. Chen Q, Tian X, Fan J, et al., An interpenetrating alginate/ gelatin network for three-dimensional (3D) cell cultures and organ bioprinting.Molecules, 25(3):756. https://doi.org/10.3390/molecules25030756 

37. Bordoni M, Karabulut E, Kuzmenko V, et al., 3D printed conductive nanocellulose scaffolds for the differentiation of human neuroblastoma cells. Cells, 9(3):682. https://doi.org/10.3390/cells9030682. 

38. Fantini V, Bordoni M, Scocozza F, et al., 2019 Bioink composition and printing parameters for 3D modeling neural tissue. Cells, 8(8):830. https://doi.org/10.3390/cells8080830 

39. Jury M, Matthiesen I, Boroojeni FR, et al., 2022, Bioorthogonally cross-linked hyaluronan-laminin hydrogels for 3D neuronal cell culture and biofabrication. Adv Healthc Mater, 11(11):e2102097. https://doi.org/10.1002/adhm.202102097 

40. Bao B, Ahmad A, Azmi AS, et al., 2013, Overview of cancer stem cells (CSCs) and mechanisms of their regulation: Implications for cancer therapy. Curr Protoc Pharmacol, Chapter 14:Unit 14 25. https://doi.org/10.1002/0471141755.ph1425s61 

41. Nothdurfter D, Ploner C, Coraca-Huber DC, et al., 2022, 3D bioprinted, vascularized neuroblastoma tumor environment in fluidic chip devices for precision medicine drug testing. Biofabrication,14(3). https://doi.org/10.1088/1758-5090/ac5fb7 

42. Ning L, Shim J, Tomov ML, et al., 2022, A 3D bioprinted in vitro model of neuroblastoma recapitulates dynamic tumor-endothelial cell interactions contributing to solid tumor aggressive behavior. Adv Sci (Weinh), 9(23):e2200244, https://doi.org/10.1002/advs.202200244 

43. Grunewald L, Lam T, Andersch L, et al., 2021, A reproducible bioprinted 3D tumor model serves as a preselection tool for CAR T cell therapy optimization. Front Immunol, 12:689697. https://doi.org/10.3389/fimmu.2021.689697 

44. Wu X, Nelson M, Basu M, et al., 2021, MYC oncogene is associated with suppression of tumor immunity and targeting Myc induces tumor cell immunogenicity for therapeutic whole cell vaccination. J Immunother Cancer, 9(3):e001388. https://doi.org/10.1136/jitc-2020-001388

Share
Back to top
International Journal of Bioprinting, Electronic ISSN: 2424-8002 Print ISSN: 2424-7723, Published by AccScience Publishing