The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines
Abstract
:1. Nanotechnology Provides Innovative Approaches to Cancer Management
2. Genetic Nanomedicines and the Main Challenges for Their Translation to the Clinic
3. Zebrafish as a Model Species
4. Zebrafish Is Currently Being Used for the Development of Anticancer Therapeutics
5. The Potential of Zebrafish for Increasing the Translation of Genetic Anticancer Nanomedicines: Barriers and Models
5.1. Toxicity
5.2. Stability and Half-Life While in Circulation
5.3. Extravasation, Penetration into the Tumor, and Interaction with the Target Cells
5.4. Functional Assays
6. Conclusions
Acknowledgments
Conflicts of Interest
References
- Swain, S.; Sahu, P.K.; Beg, S.; Babu, S.M. Nanoparticles for Cancer Targeting: Current and Future Directions. Curr. Drug Deliv. 2016, 13, 1290–1302. [Google Scholar] [CrossRef] [PubMed]
- Nascimento, T.L.; Hillaireau, H.; Vergnaud, J.; Fattal, E. Lipid-based nanosystems for CD44 targeting in cancer treatment: Recent significant advances, ongoing challenges and unmet needs. Nanomedicine 2016, 11, 1865–1887. [Google Scholar] [CrossRef] [PubMed]
- Thotakura, N.; Dadarwal, M.; Kumar, R.; Singh, B.; Sharma, G.; Kumar, P.; Katare, O.P.; Raza, K. Chitosan-palmitic acid based polymeric micelles as promising carrier for circumventing pharmacokinetic and drug delivery concerns of tamoxifen. Int. J. Biol. Macromol. 2017, 102, 1220–1225. [Google Scholar] [CrossRef] [PubMed]
- Tang, M.; Hu, P.; Zheng, Q.; Tirelli, N.; Yang, X.; Wang, Z.; Wang, Y.; Tang, Q.; He, Y. Polymeric micelles with dual thermal and reactive oxygen species (ROS)-responsiveness for inflammatory cancer cell delivery. J. Nanobiotechnol. 2017, 15, 39. [Google Scholar] [CrossRef] [PubMed]
- Song, W.; Ma, Z.; Zhang, Y.; Yang, C. Autophagy plays a dual role during intracellular siRNA delivery by lipoplex and polyplex nanoparticles. Acta Biomater. 2017, 58, 196–204. [Google Scholar] [CrossRef] [PubMed]
- Sepúlveda-Crespo, D.; Jiménez, J.L.; Gómez, R.; De La Mata, F.J.; Majano, P.L.; Muñoz-Fernández, M.Á.; Gastaminza, P. Polyanionic carbosilane dendrimers prevent hepatitis C virus infection in cell culture. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 49–58. [Google Scholar] [CrossRef] [PubMed]
- Zhong, Y.; Jia, T.; Sun, Z.; Lu, Y.; Gao, J.; Zou, H.; Xie, F.; Xu, H.; Sun, D.; Yu, Y.; Zhang, G. A dual brain-targeting curcumin-loaded polymersomes ameliorated cognitive dysfunction in intrahippocampal amyloid-β1–42-injected mice. Int. J. Nanomed. 2016, 11, 3765–3775. [Google Scholar] [CrossRef] [PubMed]
- Almhanna, K.; Wright, D.; Mercade, T.M.; Van Laethem, J.-L.; Gracian, A.C.; Guillen-Ponce, C.; Faris, J.; Lopez, C.M.; Hubner, R.A.; Bendell, J.; et al. A phase II study of antibody-drug conjugate, TAK-264 (MLN0264) in previously treated patients with advanced or metastatic pancreatic adenocarcinoma expressing guanylyl cyclase C. Investig. New Drugs 2017, 35, 634–641. [Google Scholar] [CrossRef] [PubMed]
- Yu, F.; Ao, M.; Zheng, X.; Li, N.; Xia, J.; Li, Y.; Li, D.; Hou, Z.; Qi, Z.; Chen, X.D. PEG–lipid–PLGA hybrid nanoparticles loaded with berberine–phospholipid complex to facilitate the oral delivery efficiency. Drug Deliv. 2017, 24, 825–833. [Google Scholar] [CrossRef] [PubMed]
- Raviña, M.; de la Fuente, M.; Correa, J.; Sousa-Herves, A.; Pinto, J.; Fernandez-Megia, E.; Riguera, R.; Sanchez, A.; Alonso, M.J. Core−Shell Dendriplexes with Sterically Induced Stoichiometry for Gene Delivery. Macromolecules 2010, 43, 6953–6961. [Google Scholar] [CrossRef]
- De la Fuente, M.; Seijo, B.; Alonso, M.J. Design of novel polysaccharidic nanostructures for gene delivery. Nanotechnology 2008, 19, 75105. [Google Scholar] [CrossRef] [PubMed]
- De la Fuente, M.; Csaba, N.; Garcia-Fuentes, M.; Alonso, M.J. Nanoparticles as protein and gene carriers to mucosal surfaces. Nanomedicine 2008, 3, 845–857. [Google Scholar] [CrossRef] [PubMed]
- Del Pozo-Rodríguez, A.; Solinís, M.Á.; Rodríguez-Gascón, A. Applications of lipid nanoparticles in gene therapy. Eur. J. Pharm. Biopharm. 2016, 109, 184–193. [Google Scholar] [CrossRef] [PubMed]
- Yin, H.; Kanasty, R.L.; Eltoukhy, A.A.; Vegas, A.J.; Dorkin, J.R.; Anderson, D.G. Non-viral vectors for gene-based therapy. Nat. Rev. Genet. 2014, 15, 541–555. [Google Scholar] [CrossRef] [PubMed]
- Shi, J.; Kantoff, P.W.; Wooster, R.; Farokhzad, O.C. Cancer nanomedicine: Progress, challenges and opportunities. Nat. Rev. Cancer 2016, 17, 20–37. [Google Scholar] [CrossRef] [PubMed]
- Saif, M.W.U.S. Food and Drug Administration approves paclitaxel protein-bound particles (Abraxane®) in combination with gemcitabine as first-line treatment of patients with metastatic pancreatic cancer. JOP 2013, 14, 686–688. [Google Scholar] [CrossRef] [PubMed]
- Wicki, A.; Witzigmann, D.; Balasubramanian, V.; Huwyler, J. Nanomedicine in cancer therapy: Challenges, opportunities, and clinical applications. J. Control. Release 2015, 200, 138–157. [Google Scholar] [CrossRef] [PubMed]
- Rodriguez, M.A.; Pytlik, R.; Kozak, T.; Chhanabhai, M.; Gascoyne, R.; Lu, B.; Deitcher, S.R.; Winter, J.N. Vincristine sulfate liposomes injection (Marqibo) in heavily pretreated patients with refractory aggressive non-Hodgkin lymphoma: Report of the pivotal phase 2 study. Cancer 2009, 115, 3475–3482. [Google Scholar] [CrossRef] [PubMed]
- DaunoXome approved. AIDS Patient Care STDS 1996, 10, 263.
- European Medicines Agency—Science Medicines Health. Available online: http://www.ema.europa.eu/ema/ (accessed on 24 November 2017).
- U.S. Food and Drug Administration Home Page. Available online: https://www.fda.gov/ (accessed on 24 November 2017).
- Barenholz, Y. Doxil®—The first FDA-approved nano-drug: Lessons learned. J. Control. Release 2012, 160, 117–134. [Google Scholar] [CrossRef] [PubMed]
- Martínez-González, R.; Estelrich, J.; Busquets, M. Liposomes Loaded with Hydrophobic Iron Oxide Nanoparticles: Suitable T2 Contrast Agents for MRI. Int. J. Mol. Sci. 2016, 17, 1209. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.; Guo, R.; Shi, X.; Allen, S.; Cao, Z.; Baker, J.R.; Wang, S.H. Modified Nanoemulsions with Iron Oxide for Magnetic Resonance Imaging. Nanomaterials 2016, 6, 223. [Google Scholar] [CrossRef] [PubMed]
- Cui, M.; Dong, Z.; Cai, H.; Huang, K.; Liu, Y.; Fang, Z.; Li, X.; Luo, Y. Folate-targeted polymeric micelles loaded with superparamagnetic iron oxide as a contrast agent for magnetic resonance imaging of a human tongue cancer cell line. Mol. Med. Rep. 2017. [Google Scholar] [CrossRef] [PubMed]
- Pratt, E.C.; Shaffer, T.M.; Grimm, J. Nanoparticles and radiotracers: Advances toward radionanomedicine. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2016, 8, 872–890. [Google Scholar] [CrossRef] [PubMed]
- Thakor, A.S.; Gambhir, S.S. Nanooncology: The future of cancer diagnosis and therapy. CA. Cancer J. Clin. 2013, 63, 395–418. [Google Scholar] [CrossRef] [PubMed]
- Park, S.; Aalipour, A.; Vermesh, O.; Yu, J.H.; Gambhir, S.S. Towards clinically translatable in vivo nanodiagnostics. Nat. Rev. Mater. 2017, 2, 17014. [Google Scholar] [CrossRef]
- Sneider, A.; VanDyke, D.; Paliwal, S.; Rai, P. Remotely Triggered Nano-Theranostics For Cancer Applications. Nanotheranostics 2017, 1, 1–22. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Zhang, W.; Zhu, G.; Xie, J.; Chen, X. Rethinking cancer nanotheranostics. Nat. Rev. Mater. 2017, 2, 17024. [Google Scholar] [CrossRef] [PubMed]
- Baetke, S.C.; Lammers, T.; Kiessling, F. Applications of nanoparticles for diagnosis and therapy of cancer. Br. J. Radiol. 2015, 88. [Google Scholar] [CrossRef] [PubMed]
- Detappe, A.; Kunjachan, S.; Rottmann, J.; Robar, J.; Tsiamas, P.; Korideck, H.; Tillement, O.; Berbeco, R. AGuIX nanoparticles as a promising platform for image-guided radiation therapy. Cancer Nanotechnol. 2015, 6, 4. [Google Scholar] [CrossRef] [PubMed]
- Kotb, S.; Detappe, A.; Lux, F.; Appaix, F.; Barbier, E.L.; Tran, V.L.; Plissonneau, M.; Gehan, H.; Lefranc, F.; Rodriguez-Lafrasse, C.; et al. Gadolinium-based nanoparticles and radiation therapy for multiple brain melanoma metastases: Proof of concept before phase I trial. Theranostics 2016, 6, 418–427. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pahle, J.; Walther, W. Vectors and strategies for nonviral cancer gene therapy. Expert Opin. Biol. Ther. 2015, 16, 443–461. [Google Scholar] [CrossRef] [PubMed]
- Zhou, Z.; Liu, X.; Zhu, D.; Wang, Y.; Zhang, Z.; Zhou, X.; Qiu, N.; Chen, X.; Shen, Y. Nonviral Cancer Gene Therapy: Delivery Cascade and Vector Nanoproperty Integration. Adv. Drug Deliv. Rev. 2017. [Google Scholar] [CrossRef] [PubMed]
- FDA Press Announcements—FDA Approval Brings First Gene Therapy to the United States; U.S. Food and Drug Administration: Silver Spring, MD, USA, 2017.
- Novartis Receives First Ever FDA Approval for a CAR-T Cell Therapy, Kymriah(TM) (CTL019), for Children and Young Adults with B-cell ALL That Is Refractory or Has Relapsed at Least Twice; Novartis: Basel, Switzerland, 2017.
- Slivac, I.; Guay, D.; Mangion, M.; Champeil, J.; Gaillet, B. Non-viral nucleic acid delivery methods. Expert Opin. Biol. Ther. 2017, 17, 105–118. [Google Scholar] [CrossRef] [PubMed]
- Bottai, G.; Truffi, M.; Corsi, F.; Santarpia, L. Progress in nonviral gene therapy for breast cancer and what comes next? Expert Opin. Biol. Ther. 2017, 17, 595–611. [Google Scholar] [CrossRef] [PubMed]
- Bayraktar, R.; Pichler, M.; Kanlikilicer, P.; Ivan, C.; Kahraman, N.; Aslan, B.; Oguztuzun, S. MicroRNA 603 acts as a tumor suppressor and inhibits triple- negative breast cancer tumorigenesis by targeting elongation factor 2 kinase. Oncotarget 2017, 8, 11641–11658. [Google Scholar] [CrossRef] [PubMed]
- Bhavsar, D.; Subramanian, K.; Sethuraman, S.; Krishnan, U.M. “Nano–in–nano” hybrid liposomes increase target specificity and gene silencing efficiency in breast cancer induced SCID mice. Eur. J. Pharm. Biopharm. 2017, 119, 96–106. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Sun, X.; Shao, R.; Xu, Y.; Gao, J.; Wenquan, L. VEGF siRNA delivered by polycation liposome- encapsulated calcium phosphate nanoparticles for tumor angiogenesis inhibition in breast cancer. Int. J. Nanomed. 2017, 12, 6075–6088. [Google Scholar] [CrossRef] [PubMed]
- Xu, F.; Liao, J.Z.; Xiang, G.Y.; Zhao, P.X.; Ye, F.; Zhao, Q.; He, X.X. MiR-101 and doxorubicin codelivered by liposomes suppressing malignant properties of hepatocellular carcinoma. Cancer Med. 2017, 6, 651–661. [Google Scholar] [CrossRef] [PubMed]
- Saavedra-Alonso, S.; Zapata-Benavides, P.; Chavez-Escamilla, A.K.; Manilla-Muñoz, E.; Zamora-Avila, D.E.; Franco-Molina, M.A.; Rodriguez-Padilla, C. WT1 shRNA delivery using transferrin-conjugated PEG liposomes in an in vivo model of melanoma. Exp. Ther. Med. 2016, 12, 3778–3784. [Google Scholar] [CrossRef] [PubMed]
- Yu, T.; Xu, B.; He, L.; Xia, S.; Chen, Y.; Zeng, J.; Liu, Y.; Li, S.; Tan, X.; Ren, K.; Yao, S.; Song, X. Pigment epithelial-derived factor gene loaded novel COOH-PEG-PLGA-COOHnanoparticles promoted tumor suppression by systemic administration. Int. J. Nanomed. 2016, 11, 743–759. [Google Scholar] [CrossRef] [PubMed]
- Luo, L.; Du, T.; Zhang, J.; Zhao, W.; Cheng, H.; Yang, Y.; Wu, Y.; Wang, C.; Men, K.; Gou, M. Efficient inhibition of ovarian cancer by degradable nanoparticle-delivered survivin T34A gene. Int. J. Nanomed. 2016, 11, 501–513. [Google Scholar] [CrossRef] [PubMed]
- Cocco, E.; Deng, Y.; Shapiro, E.M.; Bortolomai, I.; Lopez, S.; Lin, K.; Bellone, S.; Cui, J.; Menderes, G.; Black, J.D.; et al. Dual-targeting nanoparticles for in vivo delivery of suicide genes to chemotherapy-resistant ovarian cancer cells. Mol. Cancer Ther. 2017, 16, 323–333. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Gao, X.; Zheng, S.; Wang, B.; Li, Y.; Zhao, C.; Muftuoglu, Y.; Chen, S.; Li, Y.; Yao, H.; Sun, H.; Mao, Q.; You, C.; Guo, G.; Wei, Y. Modified nanoparticle mediated IL-12 immunogene therapy for colon cancer. Nanomedicine 2017. [Google Scholar] [CrossRef] [PubMed]
- Li, L.; Li, X.; Wu, Y.; Song, L.; Yang, X.; He, T.; Wang, N.; Yang, S.; Zeng, Y.; Wu, Q.; et al. Multifunctional Nucleus-targeting Nanoparticles with Ultra-high Gene Transfection Efficiency for In Vivo Gene Therapy. Theranostics 2017, 7, 1633–1649. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.B.; Zhang, K.; Tam, Y.Y.C.; Quick, J.; Tam, Y.K.; Lin, P.J.; Chen, S.; Liu, Y.; Nair, J.K.; Zlatev, I.; et al. A Glu-urea-Lys Ligand-conjugated Lipid Nanoparticle/siRNA System Inhibits Androgen Receptor Expression In Vivo. Mol. Ther. Nucleic Acids 2016, 5, e348. [Google Scholar] [CrossRef] [PubMed]
- D’Abundo, L.; Callegari, E.; Bresin, A.; Chillemi, A.; Elamin, B.K.; Guerriero, P.; Huang, X.; Saccenti, E.; Hussein, E.M.A.A.; Casciano, F.; et al. Anti-leukemic activity of microRNA-26a in a chronic lymphocytic leukemia mouse model. Oncogene 2017, 1–10. [Google Scholar] [CrossRef] [PubMed]
- De la Fuente, M.; Jones, M.C.; Santander-Ortega, M.J.; Mirenska, A.; Marimuthu, P.; Uchegbu, I.; Schätzlein, A. A nano-enabled cancer-specific ITCH RNAi chemotherapy booster for pancreatic cancer. Nanomed. Nanotechnol. Biol. Med. 2015, 11, 369–377. [Google Scholar] [CrossRef] [PubMed]
- Ramamoorth, M.; Narvekar, A. Non viral vectors in gene therapy—An overview. J. Clin. Diagnostic Res. 2015, 9, GE01–GE06. [Google Scholar] [CrossRef] [PubMed]
- Home—ClinicalTrials.gov. Available online: https://clinicaltrials.gov/ (accessed on 24 November 2017).
- Shanker, M.; Jin, J.; Branch, C.D.; Miyamoto, S.; Grimm, E.A.; Roth, J.A.; Ramesh, R. Tumor suppressor gene-based nanotherapy: From test tube to the clinic. J. Drug Deliv. 2011, 2011, 465845. [Google Scholar] [CrossRef] [PubMed]
- Dowdy, S.F. Overcoming cellular barriers for RNA therapeutics. Nat. Biotechnol. 2017, 35, 222–229. [Google Scholar] [CrossRef] [PubMed]
- McErlean, E.M.; McCrudden, C.M.; McCarthy, H.O. Delivery of nucleic acids for cancer gene therapy: Overcoming extra- and intra- cellular barriers. Ther. Deliv. 2016, 7, 619–637. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Jiang, Y.; Peng, H.; Chen, Y.; Zhu, P.; Huang, Y. Recent progress in microRNA delivery for cancer therapy by non-viral synthetic vectors. Adv. Drug Deliv. Rev. 2015, 81, 142–160. [Google Scholar] [CrossRef] [PubMed]
- Santander-Ortega, M.J.; de la Fuente, M.; Lozano, M.V.; Bekheet, M.E.; Progatzky, F.; Elouzi, A.; Uchegbu, I.F.; Schätzlein, A.G. Hydration forces as a tool for the optimization of core–shell nanoparticle vectors for cancer gene therapy. Soft Matter 2012, 8, 12080. [Google Scholar] [CrossRef]
- Santander-Ortega, M.J.; de la Fuente, M.; Lozano, M.V.; Tsui, M.L.; Bolton, K.; Uchegbu, I.F.; Schätzlein, A.G. Optimisation of synthetic vector systems for cancer gene therapy—The role of the excess of cationic dendrimer under physiological conditions. Curr. Top. Med. Chem. 2014, 14, 1172–1181. [Google Scholar] [CrossRef] [PubMed]
- De la Fuente, M.; Raviña, M.; Sousa-Herves, A.; Correa, J.; Riguera, R.; Fernandez-Megia, E.; Sánchez, A.; Alonso, M.J. Exploring the efficiency of gallic acid-based dendrimers and their block copolymers with PEG as gene carriers. Nanomedicine 2012, 7, 1667–1681. [Google Scholar] [CrossRef] [PubMed]
- Lieschke, G.J.; Currie, P.D. Animal models of human disease: Zebrafish swim into view. Nat. Rev. Genet. 2007, 8, 353–367. [Google Scholar] [CrossRef] [PubMed]
- Santoriello, C.; Zon, L.I. Science in medicine Hooked! Modeling human disease in zebrafish. J. Clin. Investig. 2012, 122, 2337–2343. [Google Scholar] [CrossRef] [PubMed]
- Ablain, J.; Zon, L.I. Of fish and men: Using zebrafish to fight human diseases. Trends Cell Biol. 2013, 23, 584–586. [Google Scholar] [CrossRef] [PubMed]
- Giannaccini, M.; Cuschieri, A.; Dente, L.; Raffa, V. Non-mammalian vertebrate embryos as models in nanomedicine. Nanomed. Nanotechnol. Biol. Med. 2014, 10, 703–719. [Google Scholar] [CrossRef] [PubMed]
- Howe, K.; Clark, M.; Torroja, C.; Torrance, J.; Berthelot, C.; Muffato, M.; Collins, J.E.; Humphray, S.; McLaren, K.; Matthews, L.; et al. The zebrafish reference genome sequence and its relationship to the human genome. Nature 2013, 496, 498–503. [Google Scholar] [CrossRef] [PubMed]
- Phillips, J.B.; Westerfield, M. Zebrafish models in translational research: Tipping the scales toward advancements in human health. Dis. Model. Mech. 2014, 7, 739–743. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.Y.; Jang, G.H.; Byun, C.H.; Jeun, M.; Searson, P.C.; Lee, K.H. Zebrafish models for functional and toxicological screening of nanoscale drug delivery systems: Promoting preclinical applications. Biosci. Rep. 2017, 37, BSR20170199. [Google Scholar] [CrossRef] [PubMed]
- Kimmel, C.B.; Ballard, W.W.; Kimmel, S.R.; Ullmann, B.; Schilling, T.F. Stages of embryonic development of the zebrafish. Dev. Dyn. 1995, 203, 253–310. [Google Scholar] [CrossRef] [PubMed]
- Cui, C.; Benard, E.L.; Kanwal, Z.; Stockhammer, O.W.; van der Vaart, M.; Zakrzewska, A.; Spaink, H.P.; Meijer, A.H. Infectious Disease Modeling and Innate Immune Function in Zebrafish Embryos, 3rd ed.; Elsevier Inc.: Amsterdam, The Netherlands, 2011; Volume 105, ISBN 9780123813206. [Google Scholar]
- Lam, S.H.; Chua, H.L.; Gong, Z.; Lam, T.J.; Sin, Y.M. Development and maturation of the immune system in zebrafish, Danio rerio: A gene expression profiling, in situ hybridization and immunological study. Dev. Comp. Immunol. 2004, 28, 9–28. [Google Scholar] [CrossRef]
- Li, F.; Zhang, S.; Wang, Z.; Li, H. Genes of the adaptive immune system are expressed early in zebrafish larval development following lipopolysaccharide stimulation. Chin. J. Oceanol. Limnol. 2011, 29, 326–333. [Google Scholar] [CrossRef]
- Bill, B.R.; Petzold, A.M.; Clark, K.J.; Schimmenti, L.A.; Ekker, S.C. A Primer for Morpholino Use in Zebrafish. Zebrafish 2009, 6, 69–77. [Google Scholar] [CrossRef] [PubMed]
- Bedell, V.M.; Westcot, S.E.; Ekker, S.C. Lessons from morpholino-based screening in zebrafish. Brief. Funct. Genom. 2011, 10, 181–188. [Google Scholar] [CrossRef] [PubMed]
- Timme-Laragy, A.R.; Karchner, S.I.; Hahn, M.E. Developmental Toxicology. Methods Mol Biol. 2012, 889, 51–71. [Google Scholar] [CrossRef] [PubMed]
- Irion, U.; Krauss, J.; Nusslein-Volhard, C. Precise and efficient genome editing in zebrafish using the CRISPR/Cas9 system. Development 2014, 141, 4827–4830. [Google Scholar] [CrossRef] [PubMed]
- Shah, A.N.; Davey, C.F.; Whitebirch, A.C.; Miller, A.C.; Moens, C.B. Rapid Reverse Genetic Screening Using CRISPR in Zebrafish. Nat. Methods 2015, 12, 535–540. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Zhao, L.; Page-McCaw, P.S.; Chen, W. Zebrafish Genome Engineering Using the CRISPR–Cas9 System. Trends Genet. 2016, 32, 815–827. [Google Scholar] [CrossRef] [PubMed]
- Albadri, S.; Del Bene, F.; Revenu, C. Genome editing using CRISPR/Cas9-based knock-in approaches in zebrafish. Methods 2017, 121–122, 77–85. [Google Scholar] [CrossRef] [PubMed]
- Haldi, M.; Ton, C.; Seng, W.L.; McGrath, P. Human melanoma cells transplanted into zebrafish proliferate, migrate, produce melanin, form masses and stimulate angiogenesis in zebrafish. Angiogenesis 2006, 9, 139–151. [Google Scholar] [CrossRef] [PubMed]
- Taylor, A.M.; Zon, L.I. Zebrafish tumor assays: The state of transplantation. Zebrafish 2009, 6, 339–346. [Google Scholar] [CrossRef] [PubMed]
- Drabsch, Y.; Snaar-Jagalska, B.E.; Ten Dijke, P. Fish tales: The use of zebrafish xenograft human cancer cell models. Histol. Histopathol. 2017, 32, 673–686. [Google Scholar] [CrossRef] [PubMed]
- Idilli, A.; Precazzini, F.; Mione, M.; Anelli, V. Zebrafish in Translational Cancer Research: Insight into Leukemia, Melanoma, Glioma and Endocrine Tumor Biology. Genes 2017, 8, 236. [Google Scholar] [CrossRef]
- Wyatt, R.A.; Trieu, N.P.V.; Crawford, B.D. Zebrafish Xenograft: An Evolutionary Experiment in Tumour Biology. Genes 2017, 8, 220. [Google Scholar] [CrossRef] [PubMed]
- Roel, M.; Rubiolo, J.A.; Botana, L.M.; Guerra-Varela, J.; Cabezas-Sainz, P.; Sanchez, L.; Silva, S.B.L.; Thomas, O.P.; Silva, S.B.L.; et al. Marine guanidine alkaloids crambescidins inhibit tumor growth and activate intrinsic apoptotic signaling inducing tumor regression in a colorectal carcinoma zebrafish xenograft model. Oncotarget 2016, 7, 83071–83087. [Google Scholar] [CrossRef] [PubMed]
- Tobin, D.M.; May, R.C.; Wheeler, R.T. Zebrafish: A see-through host and a fluorescent toolbox to probe host-pathogen interaction. PLoS Pathog. 2012, 8. [Google Scholar] [CrossRef] [PubMed]
- Fenaroli, F.; Westmoreland, D.; Benjaminsen, J.; Kolstad, T.; Skjeldal, F.M.; Meijer, A.H.; Van Der Vaart, M.; Ulanova, L.; Roos, N. Nanoparticles as Drug Delivery System against Tuberculosis in Zebrafish Embryos: Direct Visualization and treatment. ACS Nano 2014, 8, 7014–7026. [Google Scholar] [CrossRef]
- Teijeiro-Valiño, C.; Yebra-Pimentel, E.; Guerra-Varela, J.; Csaba, N.; Alonso, M.J.; Sánchez, L. Assessment of the permeability and toxicity of polymeric nanocapsules using the zebrafish model. Nanomedicine 2017, 12, 2069–2082. [Google Scholar] [CrossRef] [PubMed]
- Liu, R.; Lin, S.; Rallo, R.; Zhao, Y.; Damoiseaux, R.; Xia, T.; Lin, S.; Nel, A.; Cohen, Y. Automated phenotype recognition for zebrafish embryo based in vivo high throughput toxicity screening of engineered nano-materials. PLoS ONE 2012, 7. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.; Lin, S.; Zhao, Y.; Nel, A.E. Zebrafish: An in vivo model for nano EHS studies. Small 2013, 9, 1608–1618. [Google Scholar] [CrossRef] [PubMed]
- White, R.M.; Sessa, A.; Burke, C.; Bowman, T.; LeBlanc, J.; Ceol, C.; Bourque, C.; Dovey, M.; Goessling, W.; Burns, C.E.; et al. Transparent Adult Zebrafish as a Tool for In Vivo Transplantation Analysis. Cell Stem Cell 2008, 2, 183–189. [Google Scholar] [CrossRef] [PubMed]
- White, R.; Rose, K.; Zon, L. Zebrafish cancer: The state of the art and the path forward. Nat. Rev. Cancer 2013, 13, 624–636. [Google Scholar] [CrossRef] [PubMed]
- Dang, M.; Henderson, R.E.; Garraway, L.A.; Zon, L.I. Long-term drug administration in the adult zebrafish using oral gavage for cancer preclinical studies. Dis. Model. Mech. 2016, 9, 811–820. [Google Scholar] [CrossRef] [PubMed]
- Tang, Q.; Moore, J.C.; Ignatius, M.S.; Tenente, I.M.; Hayes, M.N.; Garcia, E.G.; Torres Yordán, N.; Bourque, C.; He, S.; Blackburn, J.S.; et al. Imaging tumour cell heterogeneity following cell transplantation into optically clear immune-deficient zebrafish. Nat. Commun. 2016, 7, 10358. [Google Scholar] [CrossRef] [PubMed]
- EFSA Opinion of the Scientific Panel on Animal Health and Welfare on a request from the Commission related to the aspects of the biology and welfare of animals used for experimental and other scientific purposes (EFSA-Q-2004-105). EFSA J. 2005, 292, 1–46.
- Russell, W.M.; Burch, R.L. The Principles of Humane Experimental Technique; Methuen Publishing: London, UK, 1959. [Google Scholar]
- Stern, H.M.; Zon, L.I. Cancer genetics and drug discovery in the zebrafish. Nat. Rev. Cancer 2003, 3, 533–539. [Google Scholar] [CrossRef] [PubMed]
- Goessling, W.; North, T.E.; Zon, L.I. New waves of discovery: Modeling cancer in zebrafish. J. Clin. Oncol. 2007, 25, 2473–2479. [Google Scholar] [CrossRef] [PubMed]
- MacRae, C.A.; Peterson, R.T. Zebrafish as tools for drug discovery. Nat. Rev. Drug Discov. 2015, 14, 721–731. [Google Scholar] [CrossRef] [PubMed]
- Deveau, A.P.; Bentley, V.L.; Berman, J.N. Using zebrafish models of leukemia to streamline drug screening and discovery. Exp. Hematol. 2017, 45, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Van Rooijen, E.; Fazio, M.; Zon, L.I. From fish bowl to bedside: The power of zebrafish to unravel melanoma pathogenesis and discover new therapeutics. Pigment Cell Melanoma Res. 2017, 30, 402–412. [Google Scholar] [CrossRef] [PubMed]
- Zhao, S.; Huang, J.; Ye, J. A fresh look at zebrafish from the perspective of cancer research. J. Exp. Clin. Cancer Res. 2015, 34, 80. [Google Scholar] [CrossRef] [PubMed]
- Lenis-Rojas, O.A.; Fernandes, A.R.; Roma-Rodrigues, C.; Baptista, P.V.; Marques, F.; Pérez-Fernández, D.; Guerra-Varela, J.; Sánchez, L.; Vázquez-García, D.; Torres, M.L.; et al. Heteroleptic mononuclear compounds of ruthenium(ii): synthesis, structural analyses, in vitro antitumor activity and in vivo toxicity on zebrafish embryos. Dalton Trans. 2016, 45, 19127–19140. [Google Scholar] [CrossRef] [PubMed]
- Lenis-Rojas, O.A.; Roma-Rodrigues, C.; Fernandes, A.R.; Marques, F.; Pérez-Fernández, D.; Guerra-Varela, J.; Sánchez, L.; Vázquez-García, D.; López-Torres, M.; Fernández, A.; et al. Dinuclear RuII(bipy)2 Derivatives: Structural, Biological, and in Vivo Zebrafish Toxicity Evaluation. Inorg. Chem. 2017, 56, 7127–7144. [Google Scholar] [CrossRef] [PubMed]
- Penas, C.; Sánchez, M.I.; Guerra-Varela, J.; Sanchez, L.; Vázquez, M.E.; Mascareñas, J.L. Light-Controlled Cellular Internalization and Cytotoxicity of Nucleic Acid-Binding Agents: Studies in Vitro and in Zebrafish Embryos. ChemBioChem 2016, 17, 37–41. [Google Scholar] [CrossRef] [PubMed]
- Blackburn, J.S.; Langenau, D.M. Zebrafish as a model to assess cancer heterogeneity, progression and relapse. Dis. Model. Mech. 2014, 7, 755–762. [Google Scholar] [CrossRef] [PubMed]
- Tat, J.; Liu, M.; Wen, X.Y. Zebrafish cancer and metastasis models for in vivo drug discovery. Drug Discov. Today Technol. 2013, 10, e83–e89. [Google Scholar] [CrossRef] [PubMed]
- Veinotte, C.J.; Dellaire, G.; Berman, J.N. Hooking the big one: The potential of zebrafish xenotransplantation to reform cancer drug screening in the genomic era. Dis. Model Mech. 2014, 7, 745–754. [Google Scholar] [CrossRef] [PubMed]
- Berghmans, S.; Butler, P.; Goldsmith, P.; Waldron, G.; Gardner, I.; Golder, Z.; Richards, F.M.; Kimber, G.; Roach, A.; Alderton, W.; et al. Zebrafish based assays for the assessment of cardiac, visual and gut function—potential safety screens for early drug discovery. J. Pharmacol. Toxicol. Methods 2008, 58, 59–68. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Kemadjou, J.R.; Zinsmeister, C.; Bauer, M.; Legradi, J.; Müller, F.; Pankratz, M.; Jäkel, J.; Strähle, U. Transcriptional profiling reveals barcode-like toxicogenomic responses in the zebrafish embryo. Genome Biol. 2007, 8, R227. [Google Scholar] [CrossRef] [PubMed]
- Simmons, S.O.; Fan, C.Y.; Ramabhadran, R. Cellular stress response pathway system as a sentinel ensemble in toxicological screening. Toxicol. Sci. 2009, 111, 202–225. [Google Scholar] [CrossRef] [PubMed]
- Barbazuk, W.B.; Korf, I.; Kadavi, C.; Heyen, J.; Tate, S.; Wun, E.; Bedell, J.A.; McPherson, J.D.; Johnson, S.L. The synthenic relationship of the zebrafish and human genomes. Genome Res. 2000, 10, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
- Huiting, L.N.; Laroche, F.; Feng, H. The Zebrafish as a Tool to Cancer Drug Discovery. Austin J. Pharmacol. Ther. 2015, 3, 1069. [Google Scholar] [PubMed]
- Lee, L.M.J.; Seftor, E.A.; Bonde, G.; Cornell, R.A.; Hendrix, M.J.C. The fate of human malignant melanoma cells transplanted into zebrafish embryos: Assessment of migration and cell division in the absence of tumor formation. Dev. Dyn. 2005, 233, 1560–1570. [Google Scholar] [CrossRef] [PubMed]
- Marques, I.J.; Weiss, F.U.; Vlecken, D.H.; Nitsche, C.; Bakkers, J.; Lagendijk, A.K.; Partecke, L.I.; Heidecke, C.-D.; Lerch, M.M.; Bagowski, C.P. Metastatic behaviour of primary human tumours in a zebrafish xenotransplantation model. BMC Cancer 2009, 9, 128. [Google Scholar] [CrossRef] [PubMed]
- Bansal, N.; Davis, S.; Tereshchenko, I.; Budak-alpdogan, T.; Zhong, H.; Stein, M.N.; Kim, I.Y.; Dipaola, R.S.; Bertino, J.R.; Sabaawy, H.E. Enrichment of human prostate cancer cells with tumor initiating properties in mouse and zebrafish xenografts by differential adhesion. Prostate 2014, 74, 187–200. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Shimada, Y.; Kuroyanagi, J.; Umemoto, N.; Nishimura, Y.; Tanaka, T. Quantitative phenotyping-based in vivo chemical screening in a zebrafish model of leukemia stem cell xenotransplantation. PLoS ONE 2014, 9, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Brown, H.K.; Schiavone, K.; Tazzyman, S.; Heymann, D.; Chico, T.J.A. Zebrafish xenograft models of cancer and metastasis for drug discovery. Expert Opin. Drug Discov. 2017, 12, 379–389. [Google Scholar] [CrossRef] [PubMed]
- Mort, R.L.; Jackson, I.J.; Patton, E.E. The melanocyte lineage in development and disease. Development 2015, 142, 1387. [Google Scholar] [CrossRef] [PubMed]
- Xie, X.; Ross, J.L.; Cowell, J.K.; Teng, Y. Future Medicinal Chemistry. Future Med. Chem. 2015, 7, 1395–1405. [Google Scholar] [CrossRef] [PubMed]
- Kim, K.-T.; Tanguay, R.L. Integrating zebrafish toxicology and nanoscience for safer product development. Green Chem. 2013, 15, 872. [Google Scholar] [CrossRef] [PubMed]
- Harper, B.; Thomas, D.; Chikkagoudar, S.; Baker, N.; Tang, K.; Heredia-Langner, A.; Lins, R.; Harper, S. Comparative hazard analysis and toxicological modeling of diverse nanomaterials using the embryonic zebrafish (EZ) metric of toxicity. J. Nanoparticle Res. 2015, 17, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Jeong, J.; Cho, H.J.; Choi, M.; Lee, W.S.; Chung, B.H.; Lee, J.S. In vivo toxicity assessment of angiogenesis and the live distribution of nano-graphene oxide and its PEGylated derivatives using the developing zebrafish embryo. Carbon N. Y. 2015, 93, 431–440. [Google Scholar] [CrossRef]
- Yang, T.; Martin, P.; Fogarty, B.; Brown, A.; Schurman, K.; Phipps, R.; Yin, V.P.; Lockman, P.; Bai, S. Exosome Delivered Anticancer Drugs Across the Blood-Brain Barrier for Brain Cancer Therapy in Danio Rerio. Pharm. Res. 2015, 32, 2003–2014. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Miao, X.; Chen, T.; Yi, X.; Wang, R.; Zhao, H.; Lee, S.M.Y.; Wang, X.; Zheng, Y. Zebrafish as a visual and dynamic model to study the transport of nanosized drug delivery systems across the biological barriers. Coll. Surf. B Biointerfaces 2017, 156, 227–235. [Google Scholar] [CrossRef] [PubMed]
- Sieber, S.; Grossen, P.; Detampel, P.; Siegfried, S.; Witzigmann, D.; Huwyler, J. Zebrafish as an early stage screening tool to study the systemic circulation of nanoparticulate drug delivery systems in vivo. J. Control. Release 2017, 264, 180–191. [Google Scholar] [CrossRef] [PubMed]
- Evensen, L.; Johansen, P.L.; Koster, G.; Zhu, K.; Herfindal, L.; Speth, M.; Fenaroli, F.; Hildahl, J.; Bagherifam, S.; Tulotta, C.; Prasmickaite, L.; Mælandsmo, G.M.; Snaar-Jagalska, E.; Griffiths, G. Zebrafish as a model system for characterization of nanoparticles against cancer. Nanoscale 2016, 8, 862–877. [Google Scholar] [CrossRef] [PubMed]
- Wehmas, L.C.; Tanguay, R.L.; Punnoose, A.; Greenwood, J.A. Developing a Novel Embryo–Larval Zebrafish Xenograft Assay to Prioritize Human Glioblastoma Therapeutics. Zebrafish 2016, 13, 317–329. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Shimada, Y.; Olsthoorn, R.C.L.; Snaar-Jagalska, B.E.; Spaink, H.P.; Kros, A. Application of Coiled Coil Peptides in Liposomal Anticancer Drug Delivery Using a Zebrafish Xenograft Model. ACS Nano 2016, 10, 7428–7435. [Google Scholar] [CrossRef] [PubMed]
- Xu, B.; Jin, Q.; Zeng, J.; Yu, T.; Chen, Y.; Li, S.; Gong, D.; He, L.; Tan, X.; Yang, L.; He, G.; Wu, J.; Song, X. Combined Tumor- and Neovascular-“Dual Targeting” Gene/Chemo-Therapy Suppresses Tumor Growth and Angiogenesis. ACS Appl. Mater. Interfaces 2016, 8, 25753–25769. [Google Scholar] [CrossRef] [PubMed]
- Aldrian, G.; Vaissière, A.; Konate, K.; Seisel, Q.; Vivès, E.; Fernandez, F.; Viguier, V.; Genevois, C.; Couillaud, F.; Démèné, H.; et al. PEGylation rate influences peptide-based nanoparticles mediated siRNA delivery in vitro and in vivo. J. Control. Release 2017, 256, 79–91. [Google Scholar] [CrossRef] [PubMed]
- Cordeiro, M.; Carvalho, L.; Silva, J.; Saúde, L.; Fernandes, A.; Baptista, P. Gold Nanobeacons for Tracking Gene Silencing in Zebrafish. Nanomaterials 2017, 7, 10. [Google Scholar] [CrossRef] [PubMed]
- Zou, D.; Wang, W.; Lei, D.; Yin, Y.; Ren, P.; Chen, J.; Yin, T.; Wang, B.; Wang, G.; Wang, Y. Penetration of blood-brain barrier and antitumor activity and nerve repair in glioma by doxorubicin-loaded monosialoganglioside micelles system. Int. J. Nanomed. 2017, 12, 4879–4889. [Google Scholar] [CrossRef] [PubMed]
- Collins, A.R.; Annangi, B.; Rubio, L.; Marcos, R.; Dorn, M.; Merker, C.; Estrela-Lopis, I.; Cimpan, M.R.; Ibrahim, M.; Cimpan, E.; et al. High throughput toxicity screening and intracellular detection of nanomaterials. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
- Brand, W.; Noorlander, C.W.; Giannakou, C.; Park, M.V.D.Z.; Vandebriel, R.J.; Bosselaers, I.E.M. Nanomedicinal products: A survey on specific toxicity and side effects. Int. J. Nanomed. 2017, 12, 6107–6129. [Google Scholar] [CrossRef] [PubMed]
- Giannakou, C.; Park, M.V.D.Z.; De Jong, W.H.; Van Loveren, H.; Vandebriel, R.J.; Geertsma, R.E. A comparison of immunotoxic effects of nanomedicinal products with regulatory immunotoxicity testing requirements. Int. J. Nanomed. 2016, 11, 2935–2952. [Google Scholar] [CrossRef] [PubMed]
- Omidi, Y.; Hollins, A.J.; Drayton, R.M.; Akhtar, S. Polypropylenimine dendrimer-induced gene expression changes: The effect of complexation with DNA, dendrimer generation and cell type. J. Drug Target. 2005, 13, 431–443. [Google Scholar] [CrossRef] [PubMed]
- Poma, A.; Di Giorgio, M.L. Toxicogenomics to improve comprehension of the mechanisms underlying responses of in vitro and in vivo systems to nanomaterials: A review. Curr. Genom. 2008, 9, 571–585. [Google Scholar] [CrossRef] [PubMed]
- Shah, V.; Taratula, O.; Garbuzenko, O.B.; Patil, M.L.; Savla, R.; Zhang, M.; Minko, T. Genotoxicity of different nanocarriers: Possible modifications for the delivery of nucleic acids. Curr. Drug Discov. Technol. 2013, 10, 8–15. [Google Scholar] [CrossRef] [PubMed]
- Anchordoquy, T.J.; Barenholz, Y.; Boraschi, D.; Chorny, M.; Decuzzi, P.; Dobrovolskaia, M.A.; Farhangrazi, Z.S.; Farrell, D.; Gabizon, A.; Ghandehari, H.; et al. Mechanisms and Barriers in Cancer Nanomedicine: Addressing Challenges, Looking for Solutions. ACS Nano 2017, 11, 12–18. [Google Scholar] [CrossRef] [PubMed]
- Guideline, T.T.; Guideline, O. OECD Guidelines for the Testing of Chemicals; Oecd/Ocde 220; Organisation for Economic Co-operation and Development: Paris, France, 2004; pp. 1–22. [Google Scholar] [CrossRef]
- Organisation for Economic Co-operation and Development (OECD). Test No. 236: Fish Embryo Acute Toxicity (FET) Test; OECD Guidelines for the Testing of Chemicals, Section 2; OECD: Paris, France, 2013; pp. 1–22. [Google Scholar] [CrossRef]
- Fent, K.; Weisbrod, C.J.; Wirth-Heller, A.; Pieles, U. Assessment of uptake and toxicity of fluorescent silica nanoparticles in zebrafish (Danio rerio) early life stages. Aquat. Toxicol. 2010, 100, 218–228. [Google Scholar] [CrossRef] [PubMed]
- Garcia, G.R.; Noyes, P.D.; Tanguay, R.L. Advancements in zebrafish applications for 21st century toxicology. Pharmacol. Ther. 2016, 161, 11–21. [Google Scholar] [CrossRef] [PubMed]
- Ong, K.J.; Zhao, X.; Thistle, M.E.; Maccormack, T.J.; Clark, R.J.; Ma, G.; Martinez-Rubi, Y.; Simard, B.; Loo, J.S.C.; Veinot, J.G.C.; Goss, G.G. Mechanistic insights into the effect of nanoparticles on zebrafish hatch. Nanotoxicology 2014, 8, 295–304. [Google Scholar] [CrossRef] [PubMed]
- Sukardi, H.; Ung, C.Y.; Gong, Z.; Lam, S.H. Incorporating zebrafish omics into chemical biology and toxicology. Zebrafish 2010, 7, 41–52. [Google Scholar] [CrossRef] [PubMed]
- Hussainzada, N.; Lewis, J.A.; Baer, C.E.; Ippolito, D.L.; Jackson, D.A.; Stallings, J.D. Whole adult organism transcriptional profiling of acute metal exposures in male zebrafish. BMC Pharmacol. Toxicol. 2014, 15, 15. [Google Scholar] [CrossRef] [PubMed]
- Geffroy, B.; Ladhar, C.; Cambier, S.; Treguer-Delapierre, M.; Brèthes, D.; Bourdineaud, J.-P. Impact of dietary gold nanoparticles in zebrafish at very low contamination pressure: The role of size, concentration and exposure time. Nanotoxicology 2012, 6, 144–160. [Google Scholar] [CrossRef] [PubMed]
- Segura-Aguilar, J.; Kostrzewa, R.M. Neurotoxins and neurotoxicity mechanisms. An overview. Neurotox. Res. 2006, 10, 263–287. [Google Scholar] [CrossRef] [PubMed]
- MacPhail, R.C.; Hunter, D.L.; Irons, T.D.; Padilla, S. Locomotion and Behavioral Toxicity in Larval Zebrafish: Background, Methods, and Data. In Zebrafish; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2011; pp. 151–164. ISBN 9781118102138. [Google Scholar]
- Truong, L.; Saili, K.S.; Miller, J.M.; Hutchison, J.E.; Tanguay, R.L. Persistent adult zebrafish behavioral deficits results from acute embryonic exposure to gold nanoparticles. Comp. Biochem. Physiol. Part C Toxicol. Pharmacol. 2012, 155, 269–274. [Google Scholar] [CrossRef] [PubMed]
- Usenko, C.Y.; Harper, S.L.; Tanguay, R.L. In vivo evaluation of carbon fullerene toxicity using embryonic zebrafish. Carbon N. Y. 2007, 45, 1891–1898. [Google Scholar] [CrossRef] [PubMed]
- Vibe, C.B.; Fenaroli, F.; Pires, D.; Wilson, S.R.; Bogoeva, V.; Kalluru, R.; Speth, M.; Anes, E.; Griffiths, G.; Hildahl, J. Thioridazine in PLGA nanoparticles reduces toxicity and improves rifampicin therapy against mycobacterial infection in zebrafish. Nanotoxicology 2016, 10, 680–688. [Google Scholar] [CrossRef] [PubMed]
- Zhuang, S.; Zhang, Z.; Zhang, W.; Bao, L.; Xu, C.; Zhang, H. Enantioselective developmental toxicity and immunotoxicity of pyraclofos toward zebrafish (Danio rerio). Aquat. Toxicol. 2015, 159, 119–126. [Google Scholar] [CrossRef] [PubMed]
- Xu, H.; Dong, X.; Zhang, Z.; Yang, M.; Wu, X.; Liu, H.; Lao, Q.; Li, C. Assessment of immunotoxicity of dibutyl phthalate using live zebrafish embryos. Fish Shellfish Immunol. 2015, 45, 286–292. [Google Scholar] [CrossRef] [PubMed]
- Daroczi, B.; Kari, G.; McAleer, M.F.; Wolf, J.C.; Rodeck, U.; Dicker, A.P. In vivo Radioprotection by the Fullerene Nanoparticle DF-1 as Assessed in a Zebrafish Model. Clin. Cancer Res. 2006, 12, 7086–7091. [Google Scholar] [CrossRef] [PubMed]
- Sheng, L.; Wang, L.; Su, M.; Zhao, X.; Hu, R.; Yu, X.; Hong, J.; Liu, D.; Xu, B.; Zhu, Y.; Wang, H.; Hong, F. Mechanism of TiO2 nanoparticle-induced neurotoxicity in zebrafish (D anio rerio). Environ. Toxicol. 2016, 31, 163–175. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Zhu, X.; Zhang, X.; Zhao, Z.; Liu, H.; George, R.; Wilson-Rawls, J.; Chang, Y.; Chen, Y. Disruption of zebrafish (Danio rerio) reproduction upon chronic exposure to TiO2 nanoparticles. Chemosphere 2011, 83, 461–467. [Google Scholar] [CrossRef] [PubMed]
- Jang, G.H.; Lee, K.Y.; Choi, J.; Kim, S.H.; Lee, K.H. Multifaceted toxicity assessment of catalyst composites in transgenic zebrafish embryos. Environ. Pollut. 2016, 216, 755–763. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.; Zhao, Y.; Xia, T.; Meng, H.; Ji, Z.; Liu, R.; George, S.; Xiong, S.; Wang, X.; Zhang, H.; et al. High content screening in zebrafish speeds up hazard ranking of transition metal oxide nanoparticles. ACS Nano 2011, 5, 7284–7295. [Google Scholar] [CrossRef] [PubMed]
- Chang, J.; Ichihara, G.; Shimada, Y.; Tada-Oikawa, S.; Kuroyanagi, J.; Zhang, B.; Suzuki, Y.; Sehsah, R.; Kato, M.; Tanaka, T.; et al. Copper Oxide Nanoparticles Reduce Vasculogenesis in Transgenic Zebrafish Through Down-Regulation of Vascular Endothelial Growth Factor Expression and Induction of Apoptosis. J. Nanosci. Nanotechnol. 2015, 15, 2140–2147. [Google Scholar] [CrossRef] [PubMed]
- Bar-Ilan, O.; Louis, K.M.; Yang, S.P.; Pedersen, J.A.; Hamers, R.J.; Peterson, R.E.; Heideman, W. Titanium dioxide nanoparticles produce phototoxicity in the developing zebrafish. Nanotoxicology 2012, 6, 670–679. [Google Scholar] [CrossRef] [PubMed]
- Zhang, W.; Lin, K.; Sun, X.; Dong, Q.; Huang, C.; Wang, H.; Guo, M.; Cui, X. Toxicological effect of MPA–CdSe QDs exposure on zebrafish embryo and larvae. Chemosphere 2012, 89, 52–59. [Google Scholar] [CrossRef] [PubMed]
- Powell, D.R.; Huttenlocher, A. Neutrophils in the Tumor Microenvironment. Trends Immunol. 2016, 37, 41–52. [Google Scholar] [CrossRef] [PubMed]
- Chambers, S.E.J.; O’Neill, C.L.; O’Doherty, T.M.; Medina, R.J.; Stitt, A.W. The role of immune-related myeloid cells in angiogenesis. Immunobiology 2013, 218, 1370–1375. [Google Scholar] [CrossRef] [PubMed]
- Renshaw, S.A.; Loynes, C.A.; Trushell, D.M.I.; Elworthy, S.; Ingham, P.W.; Whyte, M.K.B. A transgenic zebrafish model of neutrophilic inflammation. Blood 2006, 108, 3976–3978. [Google Scholar] [CrossRef] [PubMed]
- Duan, J.; Hu, H.; Li, Q.; Jiang, L.; Zou, Y.; Wang, Y.; Sun, Z. Combined toxicity of silica nanoparticles and methylmercury on cardiovascular system in zebrafish (Danio rerio) embryos. Environ. Toxicol. Pharmacol. 2016, 44, 120–127. [Google Scholar] [CrossRef] [PubMed]
- Boushehri, M.A.S.; Lamprecht, A. Nanoparticles as drug carriers: Current issues with in vitro testing. Nanomedicine 2015, 10, 3213–3230. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, V.H.; Lee, B.J. Protein corona: A new approach for nanomedicine design. Int. J. Nanomed. 2017, 12, 3137–3151. [Google Scholar] [CrossRef] [PubMed]
- Jain, P.; Pawar, R.S.; Pandey, R.S.; Madan, J.; Pawar, S.; Lakshmi, P.K.; Sudheesh, M.S. In-vitro in-vivo correlation (IVIVC) in nanomedicine: Is protein corona the missing link? Biotechnol. Adv. 2017, 35, 889–904. [Google Scholar] [CrossRef] [PubMed]
- Harper, S.; Maddux, B.L.S.; Hutchison, J.E.; Usenko, C.; Tanguay, R. Biodistribution and Toxicity of Nanomaterials In Vivo: Effects of Composition, Size, Surface Functionalization and Route of Exposure. Nanotech 2007, 2, 666–669. [Google Scholar]
- Wilhelm, S.; Tavares, A.J.; Dai, Q.; Ohta, S.; Audet, J.; Dvorak, H.F.; Chan, W.C.W. Analysis of nanoparticle delivery to tumours. Nat. Rev. Mater. 2016, 1. [Google Scholar] [CrossRef]
- Torrice, M. Does Nanomedicine Have a Delivery Problem? ACS Publ. 2016. [Google Scholar] [CrossRef] [PubMed]
- Muntimadugu, E.; Kommineni, N.; Khan, W. Exploring the Potential of Nanotherapeutics in Targeting Tumor Microenvironment for Cancer Therapy. Pharmacol. Res. 2017, 1–14. [Google Scholar] [CrossRef] [PubMed]
- Stylianopoulos, T.; Jain, R.K. Design considerations for nanotherapeutics in oncology. Nanomedicine 2015, 11, 1893–1907. [Google Scholar] [CrossRef] [PubMed]
- Kuninty, P.R.; Schnittert, J.; Storm, G.; Prakash, J. MicroRNA Targeting to Modulate Tumor Microenvironment. Front. Oncol. 2016, 6, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Kirchberger, S.; Sturtzel, C.; Pascoal, S.; Distel, M. Quo natas, Danio?—Recent Progress in Modeling Cancer in Zebrafish. Front. Oncol. 2017, 7. [Google Scholar] [CrossRef] [PubMed]
- Wang, J.; Cao, Z.; Zhang, X.M.; Nakamura, M.; Sun, M.; Hartman, J.; Harris, R.A.; Sun, Y.; Cao, Y. Novel mechanism of macrophage-mediated metastasis revealed in a zebrafish model of tumor development. Cancer Res. 2015, 75, 306–315. [Google Scholar] [CrossRef] [PubMed]
- Rouhi, P.; Jensen, L.D.; Cao, Z.; Hosaka, K.; Länne, T.; Wahlberg, E.; Steffensen, J.F.; Cao, Y. Hypoxia-induced metastasis model in embryonic zebrafish. Nat. Protoc. 2010, 5, 1911–1918. [Google Scholar] [CrossRef] [PubMed]
- Stoletov, K.; Montel, V.; Lester, R.D.; Gonias, S.L.; Klemke, R. High-resolution imaging of the dynamic tumor cell vascular interface in transparent zebrafish. Proc. Natl. Acad. Sci. USA 2007, 104, 17406–17411. [Google Scholar] [CrossRef] [PubMed]
- Van der Ent, W.; Burrello, C.; de Lange, M.J.; van der Velden, P.A.; Jochemsen, A.G.; Jager, M.J.; Snaar-Jagalska, B.E. Embryonic Zebrafish: Different Phenotypes after Injection of Human Uveal Melanoma Cells. Ocul. Oncol. Pathol. 2015, 1, 170–181. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Shimada, Y.; Kuroyanagi, J.; Nishimura, Y.; Umemoto, N.; Nomoto, T.; Shintou, T.; Miyazaki, T.; Tanaka, T. Zebrafish xenotransplantation model for cancer stem-like cell study and high-throughput screening of inhibitors. Tumor Biol. 2014, 35, 11861–11869. [Google Scholar] [CrossRef] [PubMed]
- Eguiara, A.; Holgado, O.; Beloqui, I.; Abalde, L.; Sanchez, Y.; Callol, C.; Martin, A.G. Xenografts in zebrafish embryos as a rapid functional assay for breast cancer stem-like cell identification. Cell Cycle 2011, 10, 3751–3757. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Huang, X.; Ding, T.W.; Gong, Z. Enhanced angiogenesis, hypoxia and neutrophil recruitment during Myc-induced liver tumorigenesis in zebrafish. Sci. Rep. 2016, 6, 31952. [Google Scholar] [CrossRef] [PubMed]
- Wagner, D.S.; Delk, N.A.; Lukianova-hleb, E.Y.; Hafner, J.H.; Lapotko, D.O. The in vivo performance of plasmonic nanobubbles as cell theranostic agents in zebrafish hosting prostate cancer xenografts. Biomaterials 2010, 31, 7567–7574. [Google Scholar] [CrossRef] [PubMed]
- Jain, A.S.; Makhija, D.T.; Goel, P.N.; Shah, S.M.; Nikam, Y.; Gude, R.P.; Jagtap, A.G.; Nagarsenker, M.S. Docetaxel in cationic lipid nanocapsules for enhanced in vivo activity. Pharm. Dev. Technol. 2016, 21, 76–85. [Google Scholar] [CrossRef] [PubMed]
- Gong, C.; Deng, S.; Wu, Q.; Xiang, M.; Wei, X.; Li, L.; Gao, X.; Wang, B.; Sun, L.; Chen, Y.; et al. Improving antiangiogenesis and anti-tumor activity of curcumin by biodegradable polymeric micelles. Biomaterials 2013, 34, 1413–1432. [Google Scholar] [CrossRef] [PubMed]
- Naber, H.P.H.; Drabsch, Y.; Snaar-Jagalska, B.E.; ten Dijke, P.; van Laar, T. Snail and Slug, key regulators of TGF-β-induced EMT, are sufficient for the induction of single-cell invasion. Biochem. Biophys. Res. Commun. 2013, 435, 58–63. [Google Scholar] [CrossRef] [PubMed]
- Yang, X.J.; Cui, W.; Gu, A.; Xu, C.; Yu, S.C.; Li, T.T.; Cui, Y.H.; Zhang, X.; Bian, X.W. A Novel Zebrafish Xenotransplantation Model for Study of Glioma Stem Cell Invasion. PLoS ONE 2013, 8, 1–9. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Lamers, G.E.M.; Beenakker, J.W.M.; Cui, C.; Ghotra, V.P.S.; Danen, E.H.J.; Meijer, A.H.; Spaink, H.P.; Snaar-Jagalska, B.E. Neutrophil-mediated experimental metastasis is enhanced by VEGFR inhibition in a zebrafish xenograft model. J. Pathol. 2012, 227, 431–445. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tobia, C.; Gariano, G.; De Sena, G.; Presta, M. Zebrafish embryo as a tool to study tumor/endothelial cell cross-talk. Biochim. Biophys. Acta 2013, 1832, 1371–1377. [Google Scholar] [CrossRef] [PubMed]
- Duan, J.; Hu, H.; Feng, L.; Yang, X.; Sun, Z. Silica nanoparticles inhibit macrophage activity and angiogenesis via VEGFR2-mediated MAPK signaling pathway in zebrafish embryos. Chemosphere 2017, 183, 483–490. [Google Scholar] [CrossRef] [PubMed]
- Van Ham, T.J.; Mapes, J.; Kokel, D.; Peterson, R.T. Live imaging of apoptotic cells in zebrafish. FASEB J. 2010, 24, 4336–4342. [Google Scholar] [CrossRef] [PubMed]
- Chakraborty, C.; Sharma, A.R.; Sharma, G.; Lee, S.-S. Zebrafish: A complete animal model to enumerate the nanoparticle toxicity. J. Nanobiotechnol. 2016, 14, 65. [Google Scholar] [CrossRef] [PubMed]
- Blechinger, S.R.; Evans, T.G.; Tang, P.T.; Kuwada, J.Y.; Warren, J.T., Jr.; Krone, P.H. The heat-inducible zebrafish hsp70 gene is expressed during normal lens development under non-stress conditions. Mech. Dev. 2002, 112, 213–215. [Google Scholar] [CrossRef]
Nanocarrier | Gene Vector | Target | Indication | Administration Route | Ref |
---|---|---|---|---|---|
Liposomes | miRNA | Restoration of oncossuppressor | Breast cancer | Tail vein | [40] |
siRNA | EpCAM silencing | Breast cancer | Tumor adjacent | [41] | |
siRNA | Anti-angiogenesis | Breast cancer | Intratumoral | [42] | |
miRNA | Restoration of oncosuppressor | Hepatocellular carcinoma | Intratumoral | [43] | |
shRNA | WT1 silencing | Melanoma | Tail vein | [44] | |
Polymeric nanoparticles | pDNA | Anti-angiogenesis | Colon cancer | Tail vein | [45] |
pDNA | Induce apoptosis | Ovarian cancer | Intraperitoneal | [46] | |
pDNA | Suicide gene therapy | Ovarian cancer | Intraperitoneal | [47] | |
pDNA | Immunotherapy | Colorectal cancer | Intratumoral | [48] | |
pDNA | Suicide gene therapy | Colon cancer | Intratumoral | [49] | |
Lipid nanoparticles | siRNA | Androgen receptor silencing | Prostate cancer | Tail vein | [50] |
miRNA | Restoration of microRNA-26a | Lymphocytic leukemia | Intraperitoneal | [51] | |
Dendrimers | si/shRNA | ITCH silencing | Pancreatic cancer | Tail vein | [52] |
Model | Features | Application | Ref |
---|---|---|---|
Wild type | From nature, with pigmentation according to sex, without fluorescence | Toxicity, biodistribution, xenograft | [194] |
Flk-1:eGFP | Fluorescent vascular system | Toxicity, biodistribution, xenograft, angiogenesis, extravasation, half-life circulation, metastasis | [107,130] |
Fli-1:eGFP | [107,127,162] | ||
Gata1:DsRed | [107] | ||
Nacre/fli1:eGFP | [163] | ||
Casper fli | Without pigmentation (transparent) and fluorescent vascular system | [91] | |
Casper | Without pigmentation (transparent) | Toxicity, biodistribution, xenograft, metastasis | [91] |
ARE:eGFP | Fluorescence of reactive oxygen species (ROS) | Toxicity | [162] |
Cmlc2:eGFP | Fluorescence in the heart | Cardiotoxicity | [167] |
Mpo:GFP | Fluorescent neutrophils | Interaction, half-life circulation, immuno response | [167] |
Mpeg1:mcherry | Fluorescent macrophages | [127] | |
Hsp70:eGFP | Fluorescence of the protein HSP70 stress product | Toxicity | [195] |
© 2017 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
Gutiérrez-Lovera, C.; Vázquez-Ríos, A.; Guerra-Varela, J.; Sánchez, L.; De la Fuente, M. The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines. Genes 2017, 8, 349. https://doi.org/10.3390/genes8120349
Gutiérrez-Lovera C, Vázquez-Ríos A, Guerra-Varela J, Sánchez L, De la Fuente M. The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines. Genes. 2017; 8(12):349. https://doi.org/10.3390/genes8120349
Chicago/Turabian StyleGutiérrez-Lovera, C, AJ Vázquez-Ríos, J Guerra-Varela, L Sánchez, and M De la Fuente. 2017. "The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines" Genes 8, no. 12: 349. https://doi.org/10.3390/genes8120349