We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine
Research Article

A visualized investigation at the atomic scale of the antitumor effect of magnetic nanomedicine on gastric cancer cells

    Xiaokang Liu‡,

    The Second Clinical Medical School of Lanzhou University, the Second Hospital of Lanzhou University, Lanzhou 730030, China

    Chinese Armed Police Force General Hospital of Gansu Province, Lanzhou 730000, China

    These authors contributed equally

    Search for more papers by this author

    ,
    Xia Deng

    Key Laboratory of Magnetism & Magnetic Materials of Ministry of Education, Lanzhou University, Lanzhou 730000, China

    These authors contributed equally

    Search for more papers by this author

    ,
    Xinghua Li

    Key Laboratory of Magnetism & Magnetic Materials of Ministry of Education, Lanzhou University, Lanzhou 730000, China

    ,
    Desheng Xue

    Key Laboratory of Magnetism & Magnetic Materials of Ministry of Education, Lanzhou University, Lanzhou 730000, China

    ,
    Haoli Zhang

    State Key Laboratory of Applied Organic Chemistry, College of Chemistry & Chemical Engineering, Lanzhou University, Lanzhou 730000, China

    ,
    Tao Liu

    The Second Clinical Medical School of Lanzhou University, the Second Hospital of Lanzhou University, Lanzhou 730030, China

    ,
    Qingfang Liu

    Key Laboratory of Magnetism & Magnetic Materials of Ministry of Education, Lanzhou University, Lanzhou 730000, China

    ,
    Nigel J Mellors

    Nano Materials Group, School of Computing, Science & Engineering, University of Salford, Greater Manchester, M5 4WT, UK

    ,
    Yumin Li

    * Authors for correspondence:

    E-mail Address: liym@lzu.edu.cn

    &

    E-mail Address: pengy@lzu.edu.cn

    The Second Clinical Medical School of Lanzhou University, the Second Hospital of Lanzhou University, Lanzhou 730030, China

    These authors contributed equally

    Search for more papers by this author

    &
    Yong Peng‡,

    * Authors for correspondence:

    E-mail Address: liym@lzu.edu.cn

    &

    E-mail Address: pengy@lzu.edu.cn

    Key Laboratory of Magnetism & Magnetic Materials of Ministry of Education, Lanzhou University, Lanzhou 730000, China

    These authors contributed equally

    Search for more papers by this author

    Published Online:https://doi.org/10.2217/nnm.13.142

    Aim: Discovering which anticancer drugs attack which organelle(s) of cancer cells is essential and significant, not only for understanding their therapeutic and adverse effects, but also to enable the development of new-generation therapeutics. Here, we show that novel Fe3O4–carboxymethyl cellulose–5-fluorouracil (Fe3O4–CMC–5FU) nanomedicine can apparently enhance the antitumor effect on gastric cancer cells, and its mechanism of killing the SGC-7901 gastric cancer cells can be directly observed at the atomic scale. Materials & methods: The novel nanomedicine was prepared using the traditional antitumor drug 5FU to chemically bond onto the functionalized Fe3O4 nanoparticles (Fe3O4–CMC–5FU nanomedicine), and then was fed into SGC-7901 gastric cancer cells. The inorganic Fe3O4 nanoparticles were used to track the distribution and antitumor effect of the nanomedicine within individual SGC-7901 gastric cancer cells. Results & discussion: Atomic-level observation and tracking the elemental distribution inside individual cells proved that the magnetic nanomedicine killed the gastric cells mainly by attacking their mitochondria. The enhanced therapeutic efficacy derives from the localized high concentration and poor mobility of the aggregated Fe3O4–CMC–5FU nanomedicine in the cytoplasm. Conclusion: A brand new mechanism of Fe3O4–CMC–5FU nanomedicine killing SGC-7901 gastric cancer cells by attacking their mitochondria was discovered, which is different from the classical mechanism utilized by traditional medicine 5FU, which kills gastric cancer cells by damaging their DNA. Our work might provide a partial solution in nanomedicines or even modern anticancer medicine for the visualized investigation of their antitumor effect.

    Original submitted 25 January 2013; Revised submitted 10 May 2013

    Papers of special note have been highlighted as: • of interest •• of considerable interest

    References

    • Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J. Clin.61(2),69–90 (2011).
    • Hartgrink HH, Jansen EP, Van Grieken NC, Van De Velde CJ. Gastric cancer. Lancet374(9688),477 (2009).• Comprehensive review of the incidence, causes, pathology and treatment of gastric cancer.
    • Ajani J, Bentrem D, Besh S et al. Gastric cancer, version 2.2013: featured updates to the NCCN Guidelines. J. Natl Compr. Canc. Netw.11(5),531–546 (2012).
    • Jemal A, Siegel R, Ward E et al. Cancer statistics, 2008. CA Cancer J. Clin.58(2),71–96 (2008).
    • Clardy J, Walsh C. Lessons from natural molecules. Nature432(7019),829–837 (2004).
    • Acuna G, Möller F, Holzmeister P, Beater S, Lalkens B, Tinnefeld P. Fluorescence enhancement at docking sites of DNA-directed self-assembled nanoantennas. Science338(6106),506–510 (2012).
    • Weissleder R, Kelly K, Sun EY, Shtatland T, Josephson L. Cell-specific targeting of nanoparticles by multivalent attachment of small molecules. Nat. Biotechnol.23(11),1418–1423 (2005).
    • Jain RK, Stylianopoulos T. Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol.7(11),653–664 (2010).
    • Lee H, Lytton-Jean AKR, Chen Y et al. Molecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNA delivery. Nat. Nanotechnol.7(6),389–393 (2012).
    • 10  Lee J-H, Jang J-T, Choi J-S et al. Exchange-coupled magnetic nanoparticles for efficient heat induction. Nat. Nanotechnol.6(7),418–422 (2011).
    • 11  Orringer DA, Koo YE, Chen T, Kopelman R, Sagher O, Philbert MA. Small solutions for big problems: the application of nanoparticles to brain tumor diagnosis and therapy. Clin. Pharmacol. Ther.85(5),531–534 (2009).
    • 12  Ghosh D, Lee Y, Thomas S et al. M13-templated magnetic nanoparticles for targeted in vivo imaging of prostate cancer. Nat. Nanotechnol.7(10),677–682 (2012).
    • 13  Amirfazli A. Nanomedicine: magnetic nanoparticles hit the target. Nat. Nanotechnol.2(8),467–468 (2007).
    • 14  Mikhaylov G, Mikac U, Magaeva AA et al. Ferri-liposomes as an MRI-visible drug-delivery system for targeting tumours and their microenvironment. Nat. Nanotechnol.6(9),594–602 (2011).
    • 15  Goymer P. Imaging: early detection for pancreatic cancer. Nat. Rev. Cancer8(6),408–409 (2008).
    • 16  Von Maltzahn G, Park J-H, Lin KY et al. Nanoparticles that communicate in vivo to amplify tumour targeting. Nat. Mater.10(7),545–552 (2011).
    • 17  Kim B, Han G, Toley BJ, Kim C-K, Rotello VM, Forbes NS. Tuning payload delivery in tumour cylindroids using gold nanoparticles. Nat. Nanotechnol.5(6),465–472 (2010).
    • 18  Park J-H, Gu L, Von Maltzahn G, Ruoslahti E, Bhatia SN, Sailor MJ. Biodegradable luminescent porous silicon nanoparticles for in vivo applications. Nat. Mater.8(4),331–336 (2009).
    • 19  Jin Y, Gao X. Plasmonic fluorescent quantum dots. Nat. Nanotechnol.4(9),571–576 (2009).
    • 20  Sena M, Gao X. Designing multifunctional quantum dots for bioimaging, detection, and drug delivery. Chem. Soc. Rev.39(11),4326–4354 (2010).
    • 21  Gao X, Cui Y, Levenson RM, Chung LWK, Nie S. In vivo cancer targeting and imaging with semiconductor quantum dots. Nat. Biotechnol.22(8),969–976 (2004).
    • 22  Chang YR, Lee HY, Chen K et al. Mass production and dynamic imaging of fluorescent nanodiamonds. Nat. Nanotechnol.3(5),284–288 (2008).
    • 23  Ma X-W, Zhao Y-L, Liang X-J. Nanodiamond delivery circumvents tumor resistance to doxorubicin. Acta Pharmacol. Sin.32(5),543–544 (2011).
    • 24  Mochalin VN, Shenderova O, Ho D, Gogotsi Y. The properties and applications of nanodiamonds. Nat. Nanotechnol.7(1),11–23 (2012).
    • 25  Fan D, Yin Z, Cheong R et al. Subcellular-resolution delivery of a cytokine through precisely manipulated nanowires. Nat. Nanotechnol.5(7),545–551 (2010).
    • 26  Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol.2(12),751–760 (2007).•• Discusses the application of nanocarriers in tumor targeting, therapy and potential challenges.
    • 27  Duncan R. Polymer conjugates as anticancer nanomedicines. Nat. Rev. Cancer6(9),688–701 (2006).
    • 28  Kim JW, Galanzha EI, Shashkov EV, Moon HM, Zharov VP. Golden carbon nanotubes as multimodal photoacoustic and photothermal high-contrast molecular agents. Nat. Nanotechnol.4(10),688–694 (2009).
    • 29  Liu Z, Cai W, He L et al.In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nat. Nanotechnol.2(1),47–52 (2006).
    • 30  Feng L, Liu Z. Graphene in biomedicine: opportunities and challenges. Nanomedicine (Lond.)6(2),317–324 (2011).
    • 31  Yang K, Zhang S, Zhang G, Sun X, Lee ST, Liu Z. Graphene in mice: ultrahigh in vivo tumor uptake and efficient photothermal therapy. Nano Lett.10(9),3318–3323 (2010).
    • 32  Yu MK, Jeong YY, Park J et al. Drug-loaded superparamagnetic iron oxide nanoparticles for combined cancer imaging and therapy in vivo. Angew. Chem. Int. Edit.47(29),5362–5365 (2008).•• Demonstrates that magnetic nanomedicine can be used to detect tumors and deliver sufficient anticancer drugs.
    • 33  Horcajada P, Chalati T, Serre C et al. Porous metal–organic-framework nanoscale carriers as a potential platform for drug delivery and imaging. Nat. Mater.9(2),172–178 (2009).
    • 34  Yang L, Peng XH, Wang YA et al. Receptor-targeted nanoparticles for in vivo imaging of breast cancer. Clin. Cancer Res.15(14),4722–4732 (2009).
    • 35  Si H-Y, Li D-P, Wang T-M et al. Improving the anti-tumor effect of genistein with a biocompatible superparamagnetic drug delivery system. J. Nanosci. Nanotechnol.10(4),2325–2331 (2010).• The first study that demonstrated that magnetic nanoparticles have an enhanced antitumor effect on gastric cancer cells.
    • 36  Weissleder R. Molecular imaging in cancer. Sci. Signal.312(5777),1168 (2006).
    • 37  Liu H, Jin L, Koh SBS et al. Atomic structure of human adenovirus by cryo-EM reveals interactions among protein networks. Science329(5995),1038–1043 (2010).
    • 38  Muller DA. Structure and bonding at the atomic scale by scanning transmission electron microscopy. Nat. Mater.8(4),263–270 (2009).
    • 39  Van Schooneveldmatti M, Gloter A, Stephan O et al. Imaging and quantifying the morphology of an organic–inorganic nanoparticle at the sub-nanometre level. Nat. Nanotechnol.5(7),538–544 (2010).•• The first study that showed that scanning transmission electron microscopy and elemental analysis techniques are powerful tools for analyzing the morphology, structure, composition and elemental distribution of organic–inorganic nanoparticles. Demonstrates that liquid scanning transmission electron microscopy can be used to image live cells.
    • 40  De Jonge N, Peckys DB, Kremers GJ, Piston DW. Electron microscopy of whole cells in liquid with nanometer resolution. Proc. Natl Acad. Sci. USA106(7),2159–2164 (2009).
    • 41  Kamaly N, Xiao Z, Valencia PM, Radovic-Moreno AF, Farokhzad OC. Targeted polymeric therapeutic nanoparticles: design, development and clinical translation. Chem. Soc. Rev.41(7),2971–3010 (2012).
    • 42  Verma A, Uzun O, Hu Y et al. Surface-structure-regulated cell-membrane penetration by monolayer-protected nanoparticles. Nat. Mater.7(7),588–595 (2008).
    • 43  Muhammad F, Guo M, Qi W et al. pH-triggered controlled drug release from mesoporous silica nanoparticles via intracelluar dissolution of ZnO nanolids. J. Am. Chem. Soc.133(23),8778–8781 (2011).
    • 44  Chauhan VP, Stylianopoulos T, Martin JD et al. Normalization of tumour blood vessels improves the delivery of nanomedicines in a size-dependent manner. Nat. Nanotechnol.7(6),383–388 (2012).
    • 45  Mourya V, Inamdar N, Tiwari A. Carboxymethyl chitosan and its applications. Adv. Mat. Lett.1(1),11–33 (2010).
    • 46  Longley D, Johnston P. 5-Fluorouracil. In: Apoptosis, Cell Signaling, and Human Diseases. Springer, NY, USA, 263–278 (2007).
    • 47  Aydin RST, Pulat M. 5-Fluorouracil encapsulated chitosan nanoparticles for pH-stimulated drug delivery: evaluation of controlled release kinetics. J. Nanomater.2012,313961 (2012).• Mainly discusses the release kinetics of 5-fluorouracil encapsulated chitosan nanoparticles.
    • 48  Harris MH, Thompson CB. The role of the BCL-2 family in the regulation of outer mitochondrial membrane permeability. Cell Death Differ.7(12),1182–1191 (2000).
    • 101  Ministry of Health of the People’s Republic of China. China public health statistical yearbook. www.moh.gov.cn/publicfiles//business/htmlfiles/zwgkzt/ptjnj/index.htm