AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (2.9 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Human CIK Cells Loaded with Gold Nanoprisms as Theranostic Platform for Targeted Photoacoustic Imaging and Enhanced Immuno-Photothermal Combined Therapy

Jingjing Zhang1Fangfang Xia1Yao Yang1Caixia Yue1Chunlei Zhang1Yuming Yang1( )Lijun Ma3Gabriel Alfranca1Yanlei Liu1Yafei Hou1Weilin Jin1Jian Ni1Jesus Martinez de la Fuente1,4Daxiang Cui1,2,3( )
Institute of Nano Biomedicine and Engineering, Shanghai Engineering Center for Intelligent Diagnosis and Treatment Instrument, Department of Instrument Science and Engineering, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
National Center for Translational Medicine, Collaborative Innovational Center for System Biology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, P. R. China
Department of tumor, Shanghai Tong Ren Hospital attached to Shanghai Jiao Tong university, 786 Yuyuan Road, Shanghai 200050, P. R. China
Instituto de Nanociencia de Aragon (INA), Universidad de Zaragoza, Zaragoza, 50018, Spain
Show Author Information

Abstract

How to enhance the therapeutic efficacy of human cytokine induced killers cell (CIK) has become a great challenge. Herein, we report for the first time that human CIK cells loaded with gold nanoprisms were successfully used for targeted photoacoustic imaging, enhanced immunotherapy and photothermal therapy of gastric cancer in vivo. Gold nanoprisms were synthesized and modified with PEG; human CIK cells were prepared and incubated with PEGylated gold nanoprisms (Au GNPrs), and then the effects of human CIK cells labeled with Au NPrs on gastric cancer MGC 803 cells were evaluated and further used for targeted photoacoustic imaging, immunotherapy and photothermal therapy of gastric cancer in vivo in mice models. Results showed that PEGylated Au NPrs could be uptaken high-efficiently by human CIK cells, resultant human CIK cells labeled with AuNPrs could inhibit the growth of gastric cancer MGC 803 cells actively by induced apoptosis and G1 phase arrest, and actively target and accumulate the tumor sites in gastric cancer-bearing nude mice. Enhanced synergistic therapeutic efficacy was demonstrated with the maximal inhibition of tumor through a combination of CIK cells-based immunotherapy for three days and then a continuous gold nanoprisms-based photothermal therapy. In conclusion, human CIK cells labeled with PEGylated Au NPrs can target gastric cancer cells in vivo, enhance immunotherapy and photothermal therapy efficacy, and have a great potential in applications such as targeted imaging, immunotherapy and photothermal therapy of gastric cancer in the near future.

References

[1]

J. Ferlay, H.R. Shin, F. Bray, et al., Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int. J. Cancer, 2010, 127: 2893-2917.

[2]

R. Siegel, D. Naishadham and A. Jemal, Cancer statistics, 2013. CA Cancer J Clin, 2013, 63: 11–30.

[3]

T. Takahashi, Y. Saikawa and Y. Kitagawa, Gastric cancer: current status of diagnosis and treatment. Cancers (Basel), 2013, 5: 48-63.

[4]

I. Proserpio, S. Rausei, S. Barzaghi, et al., Multimodal treatment of gastric cancer. World journal of gastrointestinal surgery, 2014, 6: 55-58.

[5]

D. Zhang, D. Fan, New insights into the mechanisms of gastric cancer multidrug resistance and future perspectives. Future Oncol, 2010, 6: 527-537.

[6]

D. Cui, L. Zhang, X.J. Yan, et al., A microarray-based gastric carcinoma prewarning system. World J Gastroenterol, 2005, 11: 1273-1282.

[7]

F.C. Schmeel, L.C. Schmeel, S.M.Gast, et al., Adoptive immunotherapy strategies with cytokine-induced killer (CIK) cells in the treatment of hematological malignancies. International journal of molecular sciences, 2014, 15: 14632-14648.

[8]

L. Gammaitoni, L. Giraudo, V. Leuci, et al., Cytokine induced killer cells effectively kill chemo-resistant melanoma cancer stem cells. Journal of translational medicine, 2015, 13(Suppl 1): O1.

[9]

E. Rettinger, S. Huenecke, V. Pfirrmann, et al., Repetitive infusions of Cytokine-induced killer (CIK) cells for treatment of impending relapse in high-risk leukemia patients after allogeneic stem cell transplantation. Blood, 2014, 124(21): 2438-2438.

[10]

D. Sangiolo, G. Mesiano, L. Gammaitoni, et al., Cytokine-induced killer cells eradicate bone and soft-tissue sarcomas. Cancer researchI, 2014, 74: 119-129.

[11]

C. Li, S. Liang, C. Zhang, et al., Allogenic dendritic cell and tumor cell fused vaccine for targeted imaging and enhanced immunotherapeutic efficacy of gastric cancer. Biomaterials, 2015, 54: 177-187.

[12]

J.K. Chan, C.A. Hamilton, M.K. Cheung, et al., Enhanced killing of primary ovarian cancer by retargeting autologous cytokine-induced killer cells with bispecific antibodies: a preclinical study. Clinical Cancer Research, 2006, 12: 1859-1867.

[13]

Z. Zhang, L.P. Wang, Z.Z. Luo, et al., Efficacy and safety of cord blood-derived cytokine-induced killer cells in treatment of patients with malignancies. Cytotherapy, 2015, 17: 1130-1138.

[14]

X. Zhao, Z. Zhang, H. Li, et al., Cytokine induced killer cell-based immunotherapies in patients with different stages of renal cell carcinoma. Cancer Letter, 2015, 362: 192-198.

[15]

C.E. Jaekel, I.H. Schmidt-Wolf, An update on new adoptive immunotherapy strategies for solid tumors with cytokine-induced killer cells. Expert Opinion on Biological Therapy, 2014, 14: 905-916.

[16]

G. Mesiano, M. Todorovic, L. Gammaitoni, et al., Cytokine-induced killer (CIK) cells as feasible and effective adoptive immunotherapy for the treatment of solid tumors. Expert Opinion on Biological Therapy, 2012, 12: 673-684.

[17]

J. Zhang, Y. Yang, H. Fu, et al., CIK cell therapy for solid tumor. Nano Biomed. Eng., 2014, 6(2): 60-66.

[18]

I. Venugopal, S. Pernal, T. Fusinatto, et al., Quantum dot conjugated magnetic nanoparticles for targeted drug delivery and imaging. Nano Biomed. Eng., 2016, 8(1): 24-38.

[19]

J.S. Xu, Y. Cao, C.Y. Xu, et al., Phase-transition perfluorocarbon nanoparticles for ultrasound molecular imaging and therapy. Nano Biomed. Eng., 2015, 7(1): 8-19.

[20]

B. Shen, Systems molecular imaging: right around the corner. Nano Biomed. Eng., 2014, 5(1): 1-5.

[21]

Y.S. Chen, D.X. Cui, Non-spherical gold nanoparticles: tumor imaging and therapy. Nano Biomed. Eng., 2013, 5(4): 153-160.

[22]

Y. Yang, J. Zhang, F. Xia, et al., Human CIK cells loaded with Au nanorods as a theranostic platform for targeted photoacoustic imaging and enhanced immunotherapy and photothermal therapy. Nanoscale Res. Lett., 2016, 11: 285

[23]

D. Cui, Y. Yang, J. Zhang, et al., Gold nanoprims as theranostic platform for enhanced drug delivery, targeted imaging, and photothermal therapy. Nanomedicine, 2016, 12: 456.

[24]

Y.L. Liu, M. Yang, J.P. Zhang, et al., Human induced pluripotent stem cells for tumor targeted delivery of gold nanorods and enhanced photothermal therapy. ACs Nano, 2016, 10: 2375-2385.

[25]

C.C. Bao, J. Conde, J. Curtin, et al., Bioresponsive antisense DNA gold nanobeacons as a hybrid in vivo theranostics platform for the inhibition of cancer cells and metastasis. Sci. Rep., 2015, 5: 12197.

[26]

D.X. Cui, C.L. Zhang, B. Liu, et al., Regression of gastric cancer by systemic injection of RNA nanoparticles carrying both ligand and siRNA. Sci. Rep., 2015, 5: 10726

[27]

J. Conde, C.C. Bao, Y.Q. Tan, et al., Dual targeted immunotherapy via in vivo delivery of biohybrid RNAi-Peptide nanoparticles to tumor-associated macrophages and cancer cells. Adv. Fun. Mater., 2015, 25: 4183-4194.

[28]

J. Lin, S.J. Wang, P. Huang, et al., Photosensitizer-loaded gold vesicles with strong plasmonic coupling effect for imgaing-guided photothermal/ photodynamic therapy. ACs Nano, 2013, 7: 5320-5329.

[29]

I.R. Corbin, J. Chen, W. Cao, et al., Enhanced cancer-targeted delivery using engineered high-density lipoprotein-based nanocarriers. Journal of Biomedical Nanotechnology, 2007, 3: 367-376.

[30]

P.Huang, L.Bao, C.Zhang, et al. Folic acid-conjugated silica-modified gold nanorods for X-ray/CT imaging-guided dual-mode radiation and photo-thermal therapy. Biomaterials, 2011, 32: 9796-9809.

[31]

J.H. Park, G. von Maltzahn, M.J. Xu, et al., Cooperative nanomaterial system to sensitize, target, and treat tumors. Proceedings of the National Academy of Sciences, 2010, 107: 981-986.

[32]

W. Lu, Q. Huang, G. Ku, et al., Photoacoustic imaging of living mouse brain vasculature using hollow gold nanospheres. Biomaterials, 2010, 31: 2617-2626.

[33]

S.J. Liang, C. Li, C.L. Zhang, et al., CD44 monoclonal antibody-conjugated gold nanostars for targeted photoacoustic imaging and plasmonic photothermal therapy of gastric cancer stem-like cells. Theranostics, 2015, 5: 970-984.

[34]

C.J. Orendorff, T.K. Sau and C.J. Murphy, Shapedependent plasmon-resonant gold nanoparticles. Small, 2006, 2: 636-639.

[35]

C. Bao, J. Conde, E. Polo, et al., A promising road with challenges: where gold nanoparticles are in translationalresearch? Nanomedicine, 2014, 9: 2353-2370.

[36]

A. Ambrosone, P. del Pino, V. Marchesano, et al., Gold nanoprisms for photothermal cell ablation in vivo. Nanomedicine (Future Medicine), 2014, 9(13): 1913-1922.

[37]

C.C. Bao, N. Beziere, P. del Pino, et al., Gold nanoprisms as optoacoustic signal nanoamplifiers for in vivo bioimaging of gastrointestinal cancers. Small, 2013, 9: 68-74.

[38]

D.P. Yang, D.X. Cui, Advances and prospects of gold nanorods. Chemistry- An Asian Journal, 2008, 3: 2010-2022.

[39]

B. Pelaz, P. del Pino, Synthesis and applications of gold nanoparticles. Frontiers of Nanoscience, 2012, 4: 3-33.

[40]

B. Pelaz, V. Grazu, A. Ibarra, et al., Tailoring the synthesis and heating ability of gold nanoprisms for bioapplications. Langmuir, 2012, 28: 8965-8970.

[41]

B.A. Mehta, S. Wolf, Two pathways of exocytosis of cytoplasmic granule contents and target cell killing by cytokine-induced CD3CD56 killer cells. Blood, 1995, 86: 3493-3499.

[42]

G.V. Hartland, G. Schatz, Virtual issue, plasmon resonances-a physical chemistry perspective. The Journal of Physical Chemistry C, 2011, 115: 15121-15123.

[43]

X. Huang, I.H. El-Sayed, W. Qian, et al., Cancer cell imaging and photothermal therapy in the near-infrared region by using gold nanorods. Journal of the American Chemical Society, 2006, 128: 2115-2120.

[44]

S. Link, M.A. El-Sayed, Shape and size dependence of radiative, non-radiative and photothermal properties of gold nanocrystals. International Reviews in Physical Chemistry, 2000, 19: 409-453.

[45]

M.A. Mackey, M.R. Ali, L.A. Austin, et al., The most effective gold nanorod size for plasmonic photothermal therapy: theory and in vitro experiments. The Journal of Physical Chemistry B, 2014, 118: 1319-1326.

[46]

S. Ye, G. Marston, J.R. McLaughlan, et al., Engineering gold nanotubes with controlled length and near-infrared absorption for theranostic applications. Advanced Functional Materials, 2015, 25: 2117-2127.

[47]

J.V. Jokerst, A.J. Cole, S.E. Bohndiek, et al., Gold nanorods combine photoacoustic and Raman imaging for detection and treatment of ovarian cancer. Proc. SPIE 8943, Photons Plus Ultrasound: Imaging and Sensing 2014. Mar. 3, 2014: 894366.

[48]

Y. Li, T. Wen, R. Zhao, et al., Localized electric field of plasmonic nanoplatform enhanced photodynamic tumor therapy. ACS nano, 2014, 8(11): 11529-11542.

[49]

N.R. Jana, L. Gearheart and C.J. Murphy, Seed-mediated growth approach for shape-controlled synthesis of spheroidal and rod-like gold nanoparticles using a surfactant template. Advanced Materials, 2001, 13: 1389.

[50]

B. Nikoobakht, M.A. El-Sayed, Preparation and growth mechanism of gold nanorods (NRs) using seed-mediated growth method. Chemistry of Materials, 2003, 15: 1957-1962.

[51]

A.M. Alkilany, L.B. Thompson, S.P. Boulos, et al., Gold nanorods: their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Advanced drug delivery reviews, 2012, 64: 190-199.

[52]

J. Park, A. Estrada, K. Sharp, et al., Two-photon-induced photoluminescence imaging of tumors using near-infrared excited gold nanoshells. Optics Express, 2008, 16: 1590-1599.

[53]

N.J. Durr, T. Larson, D.K. Smith, et al., Two-photon luminescence imaging of cancer cells using molecularly targeted gold nanorods. Nano letters, 2007, 7: 941-945.

[54]

H. Wang, T.B. Huff, D.A. Zweifel, et al., In vitro and in vivo two-photon luminescence imaging of single gold nanorods. Proceedings of the National Academy of Sciences of the United States of America, 2005, 102: 15752-15756.

[55]

C.E. Jäkel, I.G. Schmidt-Wolf, An update on new adoptive immunotherapy strategies for solid tumors with cytokine-induced killer cells. Expert opinion on biological therapy, 2014, 14: 905-916.

[56]

R. Nishimura, J. Baker, A. Beilhack, et al., In vivo trafficking and survival of cytokine-induced killer cells resulting in minimal GVHD with retention of antitumor activity. Blood, 2008, 112: 2563-2574.

[57]

C.L. Zhang, C. Li, Y.L. Liu, et al., Gold nanoclusters-based nanoprobes for simultaneous fluorescence imaging and targeted photodynamic therapy with superior penetration and retention behavior in tumors. Adv. Fun. Mater, 2015, 25: 1314-1325.

[58]

Z.M. Li, P. Huang, X.J. Zhang, et al., RGD-conjugated dendrimer-modified gold nanorods for in vivo tumor targeting and photothermal therapy. Molcular Pharmaceutics, 2009, 7: 94-104.

[59]

J. Ruan, J.J. Ji, H. Song, et al., Fluorescent magnetic nanoparticles-labeled mesenchymal stem cells for targeted imaging and hyperthermia therapy of in vivo gastric cancer. Nanoscale Research Letters, 2012, 7: 309.

[60]

M.R. Verneris, M. Kurnacker, V. Mailander, et al., Resistence of ex vivo expanded CD3+CD56+ T cells to Fas-mediated apoptosis. CancerImmunol Immunolther, 2000, 49: 335-345.

[61]

M.R.Alderson, T.W. Tough, et.al., Fas ligand mediates activation-induced cell death in human T lymphocytes. Exp Med, 2012, 181: 71

[62]

G. Dranoff, Cytokines in cancer pathogenesis and cancer therapy. Nature Reviews Cancer, 2011, 4: 11-22

[63]

S. Thanendrarajan, M. Nowak, H. Abeken, et al., Combining cytokine induced killer cells with vaccination in cancer immunotherapy: more than one plus one? Leuk Res., 2011, 35: 1136-1142.

Nano Biomedicine and Engineering
Pages 112-127
Cite this article:
Zhang J, Xia F, Yang Y, et al. Human CIK Cells Loaded with Gold Nanoprisms as Theranostic Platform for Targeted Photoacoustic Imaging and Enhanced Immuno-Photothermal Combined Therapy. Nano Biomedicine and Engineering, 2016, 8(3): 112-127. https://doi.org/10.5101/nbe.v8i3.p112-127

354

Views

10

Downloads

6

Crossref

11

Scopus

Altmetrics

Received: 29 June 2016
Accepted: 04 July 2016
Published: 11 July 2016
© 2016 Jingjing Zhang, Fangfang Xia, Yao Yang, Caixia Yue, Chunlei Zhang, Yuming Yang, Lijun Ma, Gabriel Alfranca, Yanlei Liu, Yafei Hou, Weilin Jin, Jian Ni, Jesus Martinez de la Fuente and Daxiang Cui.

This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Return