全部 标题 作者
关键词 摘要

OALib Journal期刊
ISSN: 2333-9721
费用:99美元

查看量下载量

相关文章

更多...
Cancers  2013 

Nanomaterials and Autophagy: New Insights in Cancer Treatment

DOI: 10.3390/cancers5010296

Keywords: nanomaterials, autophagy, cancer, cancer therapy, epigenetic factors

Full-Text   Cite this paper   Add to My Lib

Abstract:

Autophagy represents a cell’s response to stress. It is an evolutionarily conserved process with diversified roles. Indeed, it controls intracellular homeostasis by degradation and/or recycling intracellular metabolic material, supplies energy, provides nutrients, eliminates cytotoxic materials and damaged proteins and organelles. Moreover, autophagy is involved in several diseases. Recent evidences support a relationship between several classes of nanomaterials and autophagy perturbation, both induction and blockade, in many biological models. In fact, the autophagic mechanism represents a common cellular response to nanomaterials. On the other hand, the dynamic nature of autophagy in cancer biology is an intriguing approach for cancer therapeutics, since during tumour development and therapy, autophagy has been reported to trigger both an early cell survival and a late cell death. The use of nanomaterials in cancer treatment to deliver chemotherapeutic drugs and target tumours is well known. Recently, autophagy modulation mediated by nanomaterials has become an appealing notion in nanomedicine therapeutics, since it can be exploited as adjuvant in chemotherapy or in the development of cancer vaccines or as a potential anti-cancer agent. Herein, we summarize the effects of nanomaterials on autophagic processes in cancer, also considering the therapeutic outcome of synergism between nanomaterials and autophagy to improve existing cancer therapies.

References

[1]  Jain, K.K. Nanotechnology in clinical laboratory diagnostics. Clin. Chim. Acta 2005, 358, 37–54, doi:10.1016/j.cccn.2005.03.014.
[2]  ASTM E2456 - 06 Standard Terminology Relating to Nanotechnology. ASTM International, 2006.
[3]  Nevozhay, D.; Kańska, U.; Budzyńska, R.; Boratyński, J. Current status of research on conjugates and related drug delivery systems in the treatment of cancer and other diseases. Postepy Hig. Med. Dosw. 2007, 61, 350–360.
[4]  Ai, J.; Biazar, E.; Jafarpour, M.; Montazeri, M.; Majdi, A.; Aminifard, S.; Zafari, M.; Akbari, H.R.; Rad, H.G. Nanotoxicology and nanoparticle safety in biomedical designs. Int. J. Nanomedicine 2011, 6, 1117–1127.
[5]  Duncan, R. The dawning era of polymer therapeutics. Nat. Rev. Drug Discov. 2003, 2, 347–360, doi:10.1038/nrd1088.
[6]  Torchilin, V.P. Recent advances with liposomes as pharmaceutical carriers. Nat. Rev. Drug Discov. 2005, 4, 145–160, doi:10.1038/nrd1632.
[7]  Blanco, E.; Kessinger, C.W.; Sumer, B.D.; Gao, J. Multifunctional micellar nanomedicine for cancer therapy. Exp. Biol. Med. 2009, 234, 123–131.
[8]  Sankhyan, A.; Pawar, P. Recent trends in noisome as vesicular drug delivery system. J. Appl. Pharm. Sci. 2012, 2, 20–32.
[9]  Sanvicens, N.; Marco, M.P. Multifunctional nanoparticles—Properties and prospects for their use in human medicine. Trends Biotechnol. 2008, 26, 425–433, doi:10.1016/j.tibtech.2008.04.005.
[10]  Svenson, S.; Tomalia, D.A. Dendrimers in biomedical applications-reflections on the field. Adv. Drug Deliv. Rev. 2005, 57, 2106–2129, doi:10.1016/j.addr.2005.09.018.
[11]  Wang, Y.; Wang, B.; Qiao, W.; Yin, T. A novel controlled release drug delivery system for multiple drugs based on electrospun nanofibers containing nanoparticles. J. Pharm Sci. 2010, 99, 4805–4811, doi:10.1002/jps.22189.
[12]  Ferrari, M. Cancer nanotechnology: Opportunities and challenges. Nat. Rev. Cancer 2005, 5, 161–171, doi:10.1038/nrc1566.
[13]  Peer, D.; Karp, J.M.; Hong, S.; Farokhzad, O.C.; Margalit, R.; Langer, R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007, 2, 751–760.
[14]  Heath, J.R.; Davis, M.E. Nanotechnology and cancer. Annu. Rev. Med. 2008, 59, 251–265, doi:10.1146/annurev.med.59.061506.185523.
[15]  Botella, P.; Abasolo, I.; Fernández, Y.; Muniesa, C.; Miranda, S.; Quesada, M.; Ruiz, J.; Schwartz, S., Jr.; Corma, A. Surface-modified silica nanoparticles for tumor-targeted delivery of camptothecin and its biological evaluation. J. Control. Release 2011, 156, 246–257, doi:10.1016/j.jconrel.2011.06.039.
[16]  Hosta, L.; Pla-Roca, M.; Arbiol, J.; López-Iglesias, C.; Samitier, J.; Cruz, L.J.; Kogan, M.J.; Albericio, F. Conjugation of Kahalalide F with gold nanoparticles to enhance in vitro antitumoral activity. Bioconjug. Chem. 2009, 20, 138–146, doi:10.1021/bc800362j.
[17]  Conte, C.; Ungaro, F.; Maglio, G.; Tirino, P.; Siracusano, G.; Sciortino, M.T.; Leone, N.; Palma, G.; Barbieri, A.; Arra, C.; et al. Biodegradable core-shell nanoassemblies for the delivery of docetaxel and Zn(II)-phthalocyanine inspired by combination therapy for cancer. J. Control. Release 2013, 10, 40–52.
[18]  Kim, I.Y.; Kang, Y.S.; Lee, D.S.; Park, H.J.; Choi, E.K.; Oh, Y.K.; Son, H.J.; Kim, J.S. Antitumor activity of EGFR targeted pH-sensitive immunoliposomes encapsulating gemcitabine in A549 xenograft nude mice. J. Control. Release 2009, 140, 55–60, doi:10.1016/j.jconrel.2009.07.005.
[19]  Strieth, S.; Eichhorn, M.E.; Werner, A.; Sauer, B.; Teifel, M.; Michaelis, U.; Berghaus, A.; Dellian, M. Paclitaxel encapsulated in cationic liposomes increases tumor microvessel leakiness and improves therapeutic efficacy in combination with Cisplatin. Clin. Cancer Res. 2008, 14, 4603–4611, doi:10.1158/1078-0432.CCR-07-4738.
[20]  Weeden, C.; Hartlieb, K.J.; Lim, L.Y. Preparation and physicochemical characterization of a novel paclitaxel-loaded amphiphilic aminocalixarene nanoparticle platform for anticancer chemotherapy. J. Pharm. Pharmacol. 2012, 64, 1403–1411, doi:10.1111/j.2042-7158.2012.01518.x.
[21]  Patil, R.; Portilla-Arias, J.; Ding, H.; Konda, B.; Rekechenetskiy, A.; Inoue, S.; Black, K.L.; Holler, E.; Ljubimova, J.Y. Cellular delivery of doxorubicin via pH-controlled hydrazone linkage using multifunctional nano vehicle based on poly(β-L-malic acid). Int. J. Mol. Sci. 2012, 13, 11681–11693, doi:10.3390/ijms130911681.
[22]  Hatakeyama, H.; Akita, H.; Ishida, E.; Hashimoto, K.; Kobayashi, H.; Aoki, T.; Yasuda, J.; Obata, K.; Kikuchi, H.; Ishida, T.; et al. Tumor targeting of doxorubicin by anti-MT1-MMP antibody-modified PEG liposomes. Int. J. Pharm. 2007, 342, 194–200, doi:10.1016/j.ijpharm.2007.04.037.
[23]  Chen, Y.; Wu, J.J.; Huang, L. Nanoparticles targeted with NGR motif deliver c-myc siRNA and doxorubicin for anticancer therapy. Mol. Ther. 2010, 18, 828–834, doi:10.1038/mt.2009.291.
[24]  Hofheinz, R.D.; Gnad-Vogt, S.U.; Beyer, U.; Hochhaus, A. Liposomal encapsulated anti-cancer drugs. Anticancer Drugs 2005, 16, 691–707, doi:10.1097/01.cad.0000167902.53039.5a.
[25]  Tripisciano, C.; Costa, S.; Kalenczuk, R.J.; Borowiak-Palen, E. Cisplatin filled multiwalled carbon nanotubes—A novel molecular hybrid of anticancer drug container. Eur. Phys. J. B 2010, 75, 141–146, doi:10.1140/epjb/e2010-00037-2.
[26]  Arsawang, U.; Saengsawang, O.; Rungrotmongkol, T.; Sornmee, P.; Wittayanarakul, K.; Remsungnen, T.; Hannongbua, S. How do carbon nanotubes serve as carriers for gemcitabine transport in a drug delivery system? J. Mol. Graph. Model. 2011, 29, 591–596, doi:10.1016/j.jmgm.2010.11.002.
[27]  Di Crescenzo, A.; Velluto, D.; Hubbell, J.A.; Fontana, A. Biocompatible dispersions of carbon nanotubes: A potential tool for intracellular transport of anticancer drugs. Nanoscale 2011, 3, 925–928, doi:10.1039/c0nr00444h.
[28]  Bhirde, A.A.; Patel, V.; Gavard, J.; Zhang, G.; Sousa, A.A.; Masedunskas, A.; Leapman, R.D.; Weigert, R.; Gutkind, J.S.; Rusling, J.F. Targeted killing of cancer cells in vivo and in vitro with EGF-directed carbon nanotube-based drug delivery. ACS Nano 2009, 3, 307–316, doi:10.1021/nn800551s.
[29]  Tong, Q.; Li, H.; Li, W.; Chen, H.; Shu, X.; Lu, X.; Wang, G. In vitro and in vivo anti-tumor effects of gemcitabine loaded with a new drug delivery system. J. Nanosci. Nanotechnol. 2011, 11, 3651–3658, doi:10.1166/jnn.2011.3804.
[30]  Gaihre, B.; Khil, M.S.; Lee, D.R.; Kim, H.Y. Gelatin-coated magnetic iron oxide nanoparticles as carrier system: Drug loading and in vitro drug release study. Int. J. Pharm. 2009, 365, 180–189, doi:10.1016/j.ijpharm.2008.08.020.
[31]  Arias, J.L.; López-Viota, M.; Delgado, A.V.; Ruiz, M.A. Iron/ethylcellulose (core/shell) nanoplatform loaded with 5-fluorouracil for cancer targeting. Colloids Surf. B Biointerfaces 2010, 77, 111–116, doi:10.1016/j.colsurfb.2010.01.030.
[32]  Wu, W.; Chen, B.; Cheng, J.; Wang, J.; Xu, W.; Liu, L.; Xia, G.; Wei, H.; Wang, X.; Yang, M.; et al. Biocompatibility of Fe3O4/DNR magnetic nanoparticles in the treatment of hematologic malignancies. Int. J. Nanomedicine 2010, 5, 1079–1084.
[33]  Yang, J.; Park, S.B.; Yoon, H.G.; Huh, Y.M.; Haam, S. Preparation of poly epsilon-caprolactone nanoparticles containing magnetite for magnetic drug carrier. Int. J. Pharm. 2006, 324, 185–190, doi:10.1016/j.ijpharm.2006.06.029.
[34]  Hua, M.Y.; Yang, H.W.; Chuang, C.K.; Tsai, R.Y.; Chen, W.J.; Chuang, K.L.; Chang, Y.H.; Chuang, H.C.; Pang, S.T. Magnetic-nanoparticle-modified paclitaxel for targeted therapy for prostate cancer. Biomaterials 2010, 31, 7355–7363, doi:10.1016/j.biomaterials.2010.05.061.
[35]  Hua, M.Y.; Liu, H.L.; Yang, H.W.; Chen, P.Y.; Tsai, R.Y.; Huang, C.Y.; Tseng, I.C.; Lyu, L.A.; Ma, C.C.; Tang, H.J.; et al. The effectiveness of a magnetic nanoparticle-based delivery system for BCNU in the treatment of gliomas. Biomaterials 2011, 32, 516–527, doi:10.1016/j.biomaterials.2010.09.065.
[36]  Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D.; et al. Photodynamic therapy of cancer: An update. CA Cancer J. Clin. 2011, 61, 250–281, doi:10.3322/caac.20114.
[37]  Verma, S.; Watt, G.M.; Mai, Z.; Hasan, T. Strategies for enhanced photodynamic therapy effects. Photochem. Photobiol. 2007, 83, 996–1005, doi:10.1111/j.1751-1097.2007.00166.x.
[38]  Markovic, Z.M.; Ristic, B.Z.; Arsikin, K.M.; Klisic, D.G.; Harhaji-Trajkovic, L.M.; Todorovic-Markovic, B.M.; Kepic, D.P.; Kravic-Stevovic, T.K.; Jovanovic, S.P.; Milenkovic, M.M.; Milivojevic, D.D.; et al. Graphene quantum dots as autophagy-inducing photodynamic agents. Biomaterials 2012, 33, 7084–7092, doi:10.1016/j.biomaterials.2012.06.060.
[39]  Lkhagvadulam, B.; Kim, J.H.; Yoon, I.; Shim, Y.K. Size-dependent photodynamic activity of gold nanoparticles conjugate of water soluble purpurin-18-N-methyl-d-glucamine. Int. J. Biomed. Biotech. 2013, Article ID 720579.
[40]  Allard, E.; Hindre, F.; Passirani, C.; Lemaire, L.; Lepareur, N.; Noiret, N.; Menei, P.; Benoit, J.P. 188Re-loaded lipid nanocapsules as a promising radiopharmaceutical carrier for internal radiotherapy of malignant gliomas. Eur. J. Nucl. Med. Mol. Imaging 2008, 35, 1838–1846, doi:10.1007/s00259-008-0735-z.
[41]  Vanpouille-Box, C.; Lacoeuille, F.; Roux, J.; Aubé, C.; Garcion, E.; Lepareur, N.; Oberti, F.; Bouchet, F.; Noiret, N.; Garin, E.; et al. Lipid nanocapsules loaded with rhenium-188 reduce tumor progression in a rat hepatocellular carcinoma model. PLoS One 2011, 6, e16926.
[42]  Hamdy, S.; Haddadi, A.; Hung, R.W.; Lavasanifar, A. Targeting dendritic cells with nano-particulate PLGA cancer vaccine formulations. Adv. Drug Deliv. Rev. 2011, 63, 943–955, doi:10.1016/j.addr.2011.05.021.
[43]  Uto, T.; Wang, X.; Sato, K.; Haraguchi, M.; Akagi, T.; Akashi, M.; Baba, M. Targeting of antigen to dendritic cells with poly(gamma-glutamic acid) nanoparticles induces antigen-specific humoral and cellular immunity. J. Immunol. 2007, 178, 2979–2986.
[44]  Uto, T.; Akagi, T.; Toyama, M.; Nishi, Y.; Shima, F.; Akashi, M.; Baba, M. Comparative activity of biodegradable nanoparticles with aluminum adjuvants: Antigen uptake by dendritic cells and induction of immune response in mice. Immunol. Lett. 2011, 140, 36–43, doi:10.1016/j.imlet.2011.06.002.
[45]  Uto, T.; Akagi, T.; Yoshinaga, K.; Toyama, M.; Akashi, M.; Baba, M. The induction of innate and adaptive immunity by biodegradable poly(γ-glutamic acid) nanoparticles via a TLR4 and MyD88 signaling pathway. Biomaterials 2011, 32, 5206–5212, doi:10.1016/j.biomaterials.2011.03.052.
[46]  Vanpouille-Box, C.; Lacoeuille, F.; Belloche, C.; Lepareur, N.; Lemaire, L.; LeJeune, J.J.; Beno?t, J.P.; Menei, P.; Couturier, O.F.; Garcion, E.; et al. Tumor eradication in rat glioma and bypass of immunosuppressive barriers using internal radiation with (188)Re-lipid nanocapsules. Biomaterials 2011, 32, 6781–6790, doi:10.1016/j.biomaterials.2011.05.067.
[47]  Somia, N.; Verma, I.M. Gene therapy: trials and tribulations. Nat. Rev. Genet. 2000, 1, 91–99, doi:10.1038/35038533.
[48]  Pichon, C.; Billiet, L.; Midoux, P. Chemical vectors for gene delivery: Uptake and intracellular trafficking. Curr. Opin. Biotechnol. 2010, 21, 640–645, doi:10.1016/j.copbio.2010.07.003.
[49]  Li, C.; Li, L.; Keates, A.C. Targeting cancer gene therapy with magnetic nanoparticles. Oncotarget 2012, 3, 365–370.
[50]  Podila, R.; Brown, J.M. Toxicity of Engineered Nanomaterials: A Physicochemical Perspective. J. Biochem. Mol. Toxicol. 2012, doi:10.1002/jbt.21442.
[51]  Li, N.; Xia, T.; Nel, A.E. The role of oxidative stress in ambient particulate matter-induced lung diseases and its implications in the toxicity of engineered nanoparticles. Free Radic. Biol. Med. 2008, 44, 1689–1699, doi:10.1016/j.freeradbiomed.2008.01.028.
[52]  Becker, H.; Herzberg, F.; Schulte, A.; Kolossa-Gehring, M. The carcinogenic potential of nanomaterials, their release from products and options for regulating them. Int. J. Hyg. Environ. Health. 2011, 214, 231–238, doi:10.1016/j.ijheh.2010.11.004.
[53]  Magaye, R.; Zhao, J.; Bowman, L.; Ding, M. Genotoxicity and carcinogenicity of cobalt-, nickel- and copper-based nanoparticles. Exp. Ther. Med. 2012, 4, 551–561.
[54]  Meng, J.; Xing, J.; Wang, Y.; Lu, J.; Zhao, Y.; Gao, X.; Wang, P.C.; Jia, L.; Liang, X. Epigenetic modulation of human breast cancer by metallofullerenol nanoparticles: In vivo treatment and in vitro analysis. Nanoscale 2011, 3, 4713–4719, doi:10.1039/c1nr10898k.
[55]  Yang, X.; Liu, J.; He, H.; Zhou, L.; Gong, C.; Wang, X.; Yang, L.; Yuan, J.; Huang, H.; He, L.; et al. SiO2 nanoparticles induce cytotoxicity and protein expression alteration in HaCaT cells. Part. Fibre Toxicol. 2010, doi:10.1186/1743-8977-7-1.
[56]  Chen, M.; von Mikecz, A. Formation of nucleoplasmic protein aggregates impairs nuclear function in response to SiO2 nanoparticles. Exp. Cell Res. 2005, 305, 51–62, doi:10.1016/j.yexcr.2004.12.021.
[57]  Gong, C.; Tao, G.; Yang, L.; Liu, J.; Liu, Q.; Zhuang, Z. SiO2 nanoparticles induce global genomic hypomethylation in HaCaT cells. Biochem. Biophys. Res. Commun. 2010, 397, 397–400, doi:10.1016/j.bbrc.2010.05.076.
[58]  Stern, S.T.; Adiseshaiah, P.P.; Crist, R.M. Autophagy and lysosomal dysfunction as emerging mechanisms of nanomaterial toxicity. Part. Fibre Toxicol. 2012, doi:10.1186/1743-8977-9-20.
[59]  Stern, S.T.; Johnson, D.N. Role for nanomaterial-autophagy interaction in neurodegenerative disease. Autophagy 2008, 4, 1097–1100.
[60]  Carew, J.S.; Nawrocki, S.T.; Cleveland, J.L. Modulating autophagy for therapeutic benefit. Autophagy 2007, 3, 464–467.
[61]  Itakura, E.; Mizushima, N. Characterization of autophagosome formation site by a hierarchical analysis of mammalian Atg proteins. Autophagy 2010, 6, 764–776, doi:10.4161/auto.6.6.12709.
[62]  Mijaljica, D.; Prescott, M.; Devenish, R.J. Microautophagy in mammalian cells: Revisiting a 40-year-old conundrum. Autophagy 2011, 7, 673–682, doi:10.4161/auto.7.7.14733.
[63]  Li, W.; Yang, Q.; Mao, Z. Chaperone-mediated autophagy: Machinery, regulation and biological consequences. Cell. Mol. Life Sci. 2011, 68, 749–763, doi:10.1007/s00018-010-0565-6.
[64]  Rabinowitz, J.D.; White, E. Autophagy and metabolism. Science 2010, 330, 1344–1348, doi:10.1126/science.1193497.
[65]  Nakatogawa, H.; Suzuki, K.; Kamada, Y.; Ohsumi, Y. Dynamics and diversity in autophagy mechanisms: lessons from yeast. Nat. Rev. Mol. Cell Biol. 2009, 10, 458–467, doi:10.1038/nrm2708.
[66]  Eskelinen, E.L.; Reggiori, F.; Baba, M.; Kovács, A.L.; Seglen, P.O. Seeing is believing: The impact of electron microscopy on autophagy research. Autophagy 2011, 7, 935–956, doi:10.4161/auto.7.9.15760.
[67]  Klionsky, D.J.; Abdalla, F.C.; Abeliovich, H.; Abraham, R.T.; Acevedo-Arozena, A.; Adeli, K.; Agholme, L.; Agnello, M.; Agostinis, P.; Aguirre-Ghiso, J.A.; et al. Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy 2012, 8, 445–544, doi:10.4161/auto.19496.
[68]  Kroemer, G.; Mari?o, G.; Levine, B. Autophagy and the integrated stress response. Mol. Cell 2010, 40, 280–293, doi:10.1016/j.molcel.2010.09.023.
[69]  Inui, M.; Martello, G.; Piccolo, S. MicroRNA control of signal transduction. Nat. Rev. Mol. Cell Biol. 2010, 11, 252–263.
[70]  Frankel, L.B.; Lund, A.H. MicroRNA regulation of autophagy. Carcinogenesis 2012, 33, 2018–2025, doi:10.1093/carcin/bgs266.
[71]  Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008, 132, 27–42, doi:10.1016/j.cell.2007.12.018.
[72]  Mathew, R.; White, E. Autophagy in tumorigenesis and energy metabolism: Friend by day, foe by night. Curr. Opin. Genet. Dev. 2011, 21, 113–119, doi:10.1016/j.gde.2010.12.008.
[73]  Yang, Z.J.; Chee, C.E.; Huang, S.; Sinicrope, F. Autophagy modulation for cancer therapy. Cancer Biol. Ther. 2011, 11, 169–176, doi:10.4161/cbt.11.2.14663.
[74]  Maiuri, M.C.; Tasdemir, E.; Criollo, A.; Morselli, E.; Vicencio, J.M.; Carnuccio, R.; Kroemer, G. Control of autophagy by oncogenes and tumor suppressor genes. Cell Death Differ. 2009, 16, 87–93, doi:10.1038/cdd.2008.131.
[75]  Zhou, S.; Zhao, L.; Kuang, M.; Zhang, B.; Liang, Z.; Yi, T.; Wei, Y.; Zhao, X. Autophagy in tumorigenesis and cancer therapy: Dr. Jekyll or Mr. Hyde? Cancer Lett. 2012, 323, 115–127, doi:10.1016/j.canlet.2012.02.017.
[76]  Hoshino, I.; Matsubara, H. MicroRNAs in cancer diagnosis and therapy: From bench to bedside. Surg. Today 2012, doi:10.1007/s00595-012-0392-5.
[77]  Wild, L.; Flanagan, J.M. Genome-wide hypomethylation in cancer may be a passive consequence of transformation. Biochim. Biophys. Acta 2010, 1806, 50–57.
[78]  Jiang, Y.; Liu, S.; Chen, X.; Cao, Y.; Tao, Y. Genome-wide distribution of DNA methylation and DNA demethylation and related chromatin regulators in cancer. Biochim. Biophys. Acta 2012, 1835, 155–163.
[79]  Feng, W.; Marquez, R.T.; Lu, Z.; Liu, J.; Lu, K.H.; Issa, J.P.; Fishman, D.M.; Yu, Y.; Bast, R.C. Imprinted tumor suppressor genes ARHI and PEG3 are the most frequently downregulated in human ovarian cancers by loss of heterozygosity and promoter methylation. Cancer 2008, 112, 1489–1502, doi:10.1002/cncr.23323.
[80]  Tschan, M.P.; Jost, M.; Batliner, J.; Fey, M.F. The autophagy gene ULK1 plays a role in AML differentiation and is negatively regulated by the oncogenic MicroRNA-106a. In 52nd Annual Meeting and Exposition, Lausanne, Switzerland, 11–13 May 2011.
[81]  Miller, T.E.; Ghoshal, K.; Ramaswamy, B.; Roy, S.; Datta, J.; Shapiro, C.L.; Jacob, S.; Majumder, S. MicroRNA-221/222 confers tamoxifen resistance in breast cancer by targeting p27Kip1. J. Biol. Chem. 2008, 283, 29897–29903.
[82]  Korkmaz, G.; le Sage, C.; Tekirdag, K.A.; Agami, R.; Gozuacik, D. miR-376b controls starvation and mTOR inhibition-related autophagy by targeting ATG4C and BECN1. Autophagy 2012, 8, 65–76.
[83]  Abraham, D.; Jackson, N.; Gundara, J.S.; Zhao, J.; Gill, A.J.; Delbridge, L.; Robinson, B.G.; Sidhu, S.B. MicroRNA profiling of sporadic and hereditary medullary thyroid cancer identifies predictors of nodal metastasis, prognosis, and potential therapeutic targets. Clin. Cancer Res. 2011, 17, 4772–4781, doi:10.1158/1078-0432.CCR-11-0242.
[84]  Zhu, H.; Wu, H.; Liu, X.; Li, B.; Chen, Y.; Ren, X.; Liu, C.G.; Yang, J.M. Regulation of autophagy by a beclin 1-targeted microRNA, miR-30a, in cancer cells. Autophagy 2009, 5, 816–823.
[85]  Frankel, L.B.; Wen, J.; Lees, M.; H?yer-Hansen, M.; Farkas, T.; Krogh, A.; J??ttel?, M.; Lund, A.H. microRNA-101 is a potent inhibitor of autophagy. EMBO J. 2011, 30, 4628–4641, doi:10.1038/emboj.2011.331.
[86]  Lisanti, M.P.; Martinez-Outschoorn, U.E.; Chiavarina, B.; Pavlides, S.; Whitaker-Menezes, D.; Tsirigos, A.; Witkiewicz, A.; Lin, Z.; Balliet, R.; Howell, A.; et al. Understanding the “lethal” drivers of tumor-stroma co-evolution: emerging role(s) for hypoxia, oxidative stress and autophagy/mitophagy in the tumor micro-environment. Cancer Biol. Ther. 2010, 10, 537–542, doi:10.4161/cbt.10.6.13370.
[87]  Morselli, E.; Galluzzi, L.; Kepp, O.; Vicencio, J.M.; Criollo, A.; Maiuri, M.C.; Kroemer, G. Anti- and pro-tumor functions of autophagy. Biochim. Biophys. Acta 2009, 1793, 1524–1532, doi:10.1016/j.bbamcr.2009.01.006.
[88]  Levine, B.; Abrams, J. p53: The Janus of autophagy? Nat. Cell Biol. 2008, 10, 637–639, doi:10.1038/ncb0608-637.
[89]  Nicholson, K.M.; Anderson, N.G. The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal. 2002, 14, 381–395, doi:10.1016/S0898-6568(01)00271-6.
[90]  Mills, K.R.; Reginato, M.; Debnath, J.; Queenan, B.; Brugge, J.S. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is required for induction of autophagy during lumen formation in vitro. Proc. Natl. Acad. Sci. USA 2004, 101, 3438–3443.
[91]  Prins, J.; Ledgerwood, E.; Ameloot, P.; Vandenabeele, P.; Faraco, P.; Bright, N.; O’Rahilly, S.; Bradley, J. Tumour necrosis factor induced autophagy and mitochondrial morphological abnormalities are mediated by TNFR-I and/or TNFR-II and do not invariably lead to cell death. Biochem. Soc. Trans. 1998, 26, S314.
[92]  Maiuri, M.C.; Criollo, A.; Kroemer, G. Crosstalk between apoptosis and autophagy within the Beclin 1 interactome. EMBO J. 2010, 29, 515–516, doi:10.1038/emboj.2009.377.
[93]  Han, J.; Hou, W.; Goldstein, L.A.; Lu, C.; Stolz, D.B.; Yin, X.M.; Rabinowich, H. Involvement of protective autophagy in TRAIL resistance of apoptosis-defective tumor cells. J. Biol. Chem. 2008, 283, 19665–19677.
[94]  Thorburn, J.; Moore, F.; Rao, A.; Barclay, W.W.; Thomas, L.R.; Grant, K.W.; Cramer, S.D.; Thorburn, A. Selective inactivation of a Fas-associated death domain protein (FADD)-dependent apoptosis and autophagy pathway in immortal epithelial cells. Mol. Biol. Cell. 2005, 16, 1189–1199, doi:10.1091/mbc.E04-10-0906.
[95]  Rikiishi, H. Novel insights into the interplay between apoptosis and autophagy. Int. J. Cell Biol. 2012, doi:10.1155/2012/317645.
[96]  Mathew, R.; Karantza-Wadsworth, V.; White, E. Role of autophagy in cancer. Nat. Rev. Cancer 2007, 7, 961–967, doi:10.1038/nrc2254.
[97]  Amaravadi, R.K.; Lippincott-Schwartz, J.; Yin, X.M.; Weiss, W.A.; Takebe, N.; Timmer, W.; DiPaola, R.S.; Lotze, M.T.; White, E. Principles and current strategies for targeting autophagy for cancer treatment. Clin. Cancer Res. 2011, 17, 654–666, doi:10.1158/1078-0432.CCR-10-2634.
[98]  Schoenlein, P.V.; Periyasamy-Thandavan, S.; Samaddar, J.S.; Jackson, W.H.; Barrett, J.T. Autophagy facilitates the progression of ERalpha-positive breast cancer cells to antiestrogen resistance. Autophagy 2009, 5, 400–403, doi:10.4161/auto.5.3.7784.
[99]  Vazquez-Martin, A.; Oliveras-Ferraros, C.; Menendez, J.A. Autophagy facilitates the development of breast cancer resistance to the anti-HER2 monoclonal antibody trastuzumab. PLoS One 2009, 4, e6251.
[100]  Milani, M.; Rzymski, T.; Mellor, H.R.; Pike, L.; Bottini, A.; Generali, D.; Harris, A.L. The role of ATF4 stabilization and autophagy in resistance of breast cancer cells treated with Bortezomib. Cancer Res. 2009, 69, 4415–4423.
[101]  Bauvy, C.; Gane, P.; Arico, S.; Codogno, P.; Ogier-Denis, E. Autophagy delays sulindac sulfide-induced apoptosis in the human intestinal colon cancer cell line HT-29. Exp. Cell Res. 2001, 268, 139–149, doi:10.1006/excr.2001.5285.
[102]  Bellodi, C.; Lidonnici, M.R.; Hamilton, A.; Helgason, G.V.; Soliera, A.R.; Ronchetti, M.; Galavotti, S.; Young, K.W.; Selmi, T.; Yacobi, R.; et al. Targeting autophagy potentiates tyrosine kinase inhibitor-induced cell death in Philadelphia chromosome-positive cells, including primary CML stem cells. J. Clin. Invest. 2009, 119, 1109–1123, doi:10.1172/JCI35660.
[103]  Gupta, A.; Roy, S.; Lazar, A.J.; Wang, W.L.; McAuliffe, J.C.; Reynoso, D.; McMahon, J.; Taguchi, T.; Floris, G.; Debiec-Rychter, M.; et al. Autophagy inhibition and antimalarials promote cell death in gastrointestinal stromal tumor (GIST). Proc. Natl. Acad. Sci. USA 2010, 107, 14333–14338, doi:10.1073/pnas.1000248107.
[104]  Shingu, T.; Fujiwara, K.; B?gler, O.; Akiyama, Y.; Moritake, K.; Shinojima, N.; Tamada, Y.; Yokoyama, T.; Kondo, S. Inhibition of autophagy at a late stage enhances imatinib-induced cytotoxicity in human malignant glioma cells. Int. J. Cancer 2009, 124, 1060–1071, doi:10.1002/ijc.24030.
[105]  Wu, Z.; Chang, P.C.; Yang, J.C.; Chu, C.Y.; Wang, L.Y.; Chen, N.T.; Ma, A.H.; Desai, S.J.; Lo, S.H.; Evans, C.P.; et al. Autophagy blockade sensitizes prostate cancer cells towards src family kinase inhibitors. Genes Cancer 2010, 1, 40–49, doi:10.1177/1947601909358324.
[106]  Carew, J.S.; Nawrocki, S.T.; Kahue, C.N.; Zhang, H.; Yang, C.; Chung, L.; Houghton, J.A.; Huang, P.; Giles, F.J.; Cleveland, J.L. Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 2007, 110, 313–322, doi:10.1182/blood-2006-10-050260.
[107]  Li, J.; Hou, N.; Faried, A.; Tsutsumi, S.; Takeuchi, T.; Kuwano, H. Inhibition of autophagy by 3-MA enhances the effect of 5-FU-induced apoptosis in colon cancer cells. Ann. Surg. Oncol. 2009, 16, 761–771, doi:10.1245/s10434-008-0260-0.
[108]  Li, J.; Hou, N.; Faried, A.; Tsutsumi, S.; Kuwano, H. Inhibition of autophagy augments 5-fluorouracil chemotherapy in human colon cancer in vitro and in vivo model. Eur. J. Cancer 2010, 46, 1900–1909, doi:10.1016/j.ejca.2010.02.021.
[109]  Qian, W.; Liu, J.; Jin, J.; Ni, W.; Xu, W. Arsenic trioxide induces not only apoptosis but also autophagic cell death in leukemia cell lines via up-regulation of Beclin-1. Leuk. Res. 2007, 31, 329–339, doi:10.1016/j.leukres.2006.06.021.
[110]  Maiuri, M.C.; Zalckvar, E.; Kimchi, A.; Kroemer, G. Self-eating and self-killing: Crosstalk between autophagy and apoptosis. Nat. Rev. Mol. Cell. Biol. 2007, 8, 741–752, doi:10.1038/nrm2239.
[111]  Kondo, Y.; Kanzawa, T.; Sawaya, R.; Kondo, S. The role of autophagy in cancer development and response to therapy. Nat. Rev. Cancer 2005, 5, 726–734, doi:10.1038/nrc1692.
[112]  Voss, V.; Senft, C.; Lang, V.; Ronellenfitsch, M.W.; Steinbach, J.P.; Seifert, V.; K?gel, D. The pan-Bcl-2 inhibitor (?)-gossypol triggers autophagic cell death in malignant glioma. Mol. Cancer Res. 2010, 8, 1002–1016, doi:10.1158/1541-7786.MCR-09-0562.
[113]  Lian, J.; Wu, X.; He, F.; Karnak, D.; Tang, W.; Meng, Y.; Xiang, D.; Ji, M.; Lawrence, T.S.; Xu, L. A natural BH3 mimetic induces autophagy in apoptosis-resistant prostate cancer via modulating Bcl-2-Beclin1 interaction at endoplasmic reticulum. Cell Death Differ. 2011, 18, 60–71, doi:10.1038/cdd.2010.74.
[114]  Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 2011, 144, 646–674, doi:10.1016/j.cell.2011.02.013.
[115]  Townsend, K.N.; Hughson, L.R.; Schlie, K.; Poon, V.I.; Westerback, A.; Lum, J.J. Autophagy inhibition in cancer therapy: Metabolic considerations for antitumor immunity. Immunol. Rev. 2012, 249, 176–194, doi:10.1111/j.1600-065X.2012.01141.x.
[116]  Wildenberg, M.E.; Vos, A.C.; Wolfkamp, S.C.; Duijvestein, M.; Verhaar, A.P.; Te Velde, A.A.; van den Brink, G.R.; Hommes, D.W. Autophagy attenuates the adaptive immune response by destabilizing the immunologic synapse. Gastroenterology 2012, 142, 1493–1503, doi:10.1053/j.gastro.2012.02.034.
[117]  Chemali, M.; Radtke, K.; Desjardins, M.; English, L. Alternative pathways for MHC class I presentation: a new function for autophagy. Cell. Mol. Life Sci. 2011, 68, 1533–1541, doi:10.1007/s00018-011-0660-3.
[118]  Schmid, D.; Pypaert, M.; Münz, C. Antigen-loading compartments for major histocompatibility complex class II molecules continuously receive input from autophagosomes. Immunity 2007, 26, 79–92, doi:10.1016/j.immuni.2006.10.018.
[119]  Gerriets, V.A.; Rathmell, J.C. Metabolic pathways in T cell fate and function. Trends Immunol. 2012, 33, 168–173, doi:10.1016/j.it.2012.01.010.
[120]  Kovacs, J.R.; Li, C.; Yang, Q.; Li, G.; Garcia, I.G.; Ju, S.; Roodman, D.G.; Windle, J.J.; Zhang, X.; Lu, B. Autophagy promotes T-cell survival through degradation of proteins of the cell death machinery. Cell Death Differ. 2012, 19, 144–152, doi:10.1038/cdd.2011.78.
[121]  Pua, H.H.; Guo, J.; Komatsu, M.; He, Y.W. Autophagy is essential for mitochondrial clearance in mature T lymphocytes. J. Immunol. 2009, 182, 4046–4055, doi:10.4049/jimmunol.0801143.
[122]  Jia, W.; He, Y.W. Temporal regulation of intracellular organelle homeostasis in T lymphocytes by autophagy. J. Immunol. 2011, 186, 5313–5322, doi:10.4049/jimmunol.1002404.
[123]  Hubbard, V.M.; Valdor, R.; Patel, B.; Singh, R.; Cuervo, A.M.; Macian, F. Macroautophagy regulates energy metabolism during effector T cell activation. J. Immunol. 2010, 185, 7349–7357, doi:10.4049/jimmunol.1000576.
[124]  Li, Y.; Wang, L.X.; Yang, G.; Hao, F.; Urba, W.J.; Hu, H.M. Efficient cross-presentation depends on autophagy in tumor cells. Cancer Res. 2008, 68, 6889–6895, doi:10.1158/0008-5472.CAN-08-0161.
[125]  Li, Y.; Wang, L.X.; Pang, P.; Cui, Z.; Aung, S.; Haley, D.; Fox, B.A.; Urba, W.J.; Hu, H.M. Tumor-derived autophagosome vaccine: Mechanism of cross-presentation and therapeutic efficacy. Clin. Cancer Res. 2011, 17, 7047–7057.
[126]  Yi, Y.; Zhou, Z.; Shu, S.; Fang, Y.; Twitty, C.; Hilton, T.L.; Aung, S.; Urba, W.J.; Fox, B.A.; Hu, H.M.; et al. Autophagy-assisted antigen cross-presentation: Autophagosome as the argo of shared tumor-specific antigens and DAMPs. Oncoimmunology 2012, 1, 976–978, doi:10.4161/onci.20059.
[127]  Kepp, O.; Galluzzi, L.; Martins, I.; Schlemmer, F.; Adjemian, S.; Michaud, M.; Sukkurwala, A.Q.; Menger, L.; Zitvogel, L.; Kroemer, G. Molecular determinants of immunogenic cell death elicited by anticancer chemotherapy. Cancer Metastasis Rev. 2011, 30, 61–69, doi:10.1007/s10555-011-9273-4.
[128]  Twitty, C.G.; Jensen, S.M.; Hu, H.M.; Fox, B.A. Tumor-derived autophagosome vaccine: induction of cross-protective immune responses against short-lived proteins through a p62-dependent mechanism. Clin. Cancer Res. 2011, 17, 6467–6481, doi:10.1158/1078-0432.CCR-11-0812.
[129]  Inguscio, V.; Panzarini, E.; Dini, L. Autophagy contributes to the death/survival balance in cancer photodynamic therapy. Cells 2012, 1, 464–491, doi:10.3390/cells1030464.
[130]  Reiners, J.J., Jr.; Agostinis, P.; Berg, K.; Oleinick, N.L.; Kessel, D. Assessing autophagy in the context of photodynamic therapy. Autophagy 2010, 6, 7–18, doi:10.4161/auto.6.1.10220.
[131]  Dewaele, M.; Maes, H.; Agostinis, P. ROS-mediated mechanisms of autophagy stimulation and their relevance in cancer therapy. Autophagy 2010, 6, 838–854, doi:10.4161/auto.6.7.12113.
[132]  Kessel, D.; Reiners, J.J., Jr. Apoptosis and autophagy after mitochondrial or endoplasmic reticulum photodamage. Photochem. Photobiol. 2007, 83, 1024–1028, doi:10.1111/j.1751-1097.2007.00088.x.
[133]  Kessel, D.; Arroyo, A.S. Apoptotic and autophagic responses to Bcl-2 inhibition and photodamage. Photochem. Photobiol. Sci. 2007, 6, 1290–1295, doi:10.1039/b707953b.
[134]  Dini, L.; Inguscio, V.; Tenuzzo, B.; Panzarini, E. Rose bengal acetate photodynamic therapy-induced autophagy. Cancer Biol. Ther. 2010, 10, 1048–1055, doi:10.4161/cbt.10.10.13371.
[135]  Panzarini, E.; Inguscio, V.; Dini, L. Timing the multiple cell death pathways initiated by Rose Bengal acetate photodynamic therapy. Cell Death Dis. 2011, 2, e169, doi:10.1038/cddis.2011.51.
[136]  Panzarini, E.; Tenuzzo, B.; Palazzo, F.; Chionna, A.; Dini, L. Apoptosis induction and mitochondria alteration in human HeLa tumor cells by photoproducts of Rose Bengal acetate. J. Photochem. Photobiol. B 2006, 83, 39–47, doi:10.1016/j.jphotobiol.2005.11.014.
[137]  Panzarini, E.; Tenuzzo, B.; Dini, L. Photodynamic therapy-induced apoptosis of HeLa cells. Ann. NY Acad. Sci. 2009, 1171, 617–626, doi:10.1111/j.1749-6632.2009.04908.x.
[138]  Kessel, D.; Castelli, M. Evidence that bcl-2 is the target of three photosensitizers that induce a rapid apoptotic response. Photochem. Photobiol. 2001, 74, 318–322, doi:10.1562/0031-8655(2001)074<0318:ETBITT>2.0.CO;2.
[139]  Weyergang, A.; Berg, K.; Kaalhus, O.; Peng, Q.; Selbo, P.K. Photodynamic therapy targets the mTOR signaling network in vitro and in vivo. Mol. Pharm. 2009, 6, 255–264.
[140]  Herd, H.L.; Malugin, A.; Ghandehari, H. Silica nanoconstruct cellular toleration threshold in vitro. J. Control. Release 2011, 153, 40–48, doi:10.1016/j.jconrel.2011.02.017.
[141]  Li, H.; Li, Y.; Jiao, J.; Hu, H.M. Alpha-alumina nanoparticles induce efficient autophagy-dependent cross-presentation and potent antitumour response. Nat. Nanotechnol. 2011, 6, 645–650.
[142]  Monick, M.M.; Powers, L.S.; Walters, K.; Lovan, N.; Zhang, M.; Gerke, A.; Hansdottir, S.; Hunninghake, G.W. Identification of an autophagy defect in smokers’ alveolar macrophages. J. Immunol. 2010, 185, 5425–5435, doi:10.4049/jimmunol.1001603.
[143]  Seleverstov, O.; Zabirnyk, O.; Zscharnack, M.; Bulavina, L.; Nowicki, M.; Heinrich, J.M.; Yezhelyev, M.; Emmrich, F.; O’Regan, R.; Bader, A. Quantum dots for human mesenchymal stem cells labeling. A size-dependent autophagy activation. Nano Lett. 2006, 6, 2826–2832, doi:10.1021/nl0619711.
[144]  Yokoyama, T.; Tam, J.; Kuroda, S.; Scott, A.W.; Aaron, J.; Larson, T.; Shanker, M.; Correa, A.M.; Kondo, S.; Roth, J.A.; et al. EGFR-targeted hybrid plasmonic magnetic nanoparticles synergistically induce autophagy and apoptosis in non-small cell lung cancer cells. PLoS One 2011, 6, e25507.
[145]  Harhaji, L.; Isakovic, A.; Raicevic, N.; Markovic, Z.; Todorovic-Markovic, B.; Nikolic, N.; Vranjes-Djuric, S.; Markovic, I.; Trajkovic, V. Multiple mechanisms underlying the anticancer action of nanocrystalline fullerene. Eur. J. Pharmacol. 2007, 568, 89–98, doi:10.1016/j.ejphar.2007.04.041.
[146]  Wu, Y.N.; Yang, L.X.; Shi, X.Y.; Li, I.C.; Biazik, J.M.; Ratinac, K.R.; Chen, D.H.; Thordarson, P.; Shieh, D.B.; Braet, F. The selective growth inhibition of oral cancer by iron core-gold shell nanoparticles through mitochondria-mediated autophagy. Biomaterials 2011, 32, 4565–4573, doi:10.1016/j.biomaterials.2011.03.006.
[147]  Khan, M.I.; Mohammad, A.; Patil, G.; Naqvi, S.A.; Chauhan, L.K.; Ahmad, I. Induction of ROS, mitochondrial damage and autophagy in lung epithelial cancer cells by iron oxide nanoparticles. Biomaterials 2012, 33, 1477–1488, doi:10.1016/j.biomaterials.2011.10.080.
[148]  Li, C.; Liu, H.; Sun, Y.; Wang, H.; Guo, F.; Rao, S.; Deng, J.; Zhang, Y.; Miao, Y.; Guo, C.; et al. PAMAM nanoparticles promote acute lung injury by inducing autophagic cell death through the Akt-TSC2-mTOR signaling pathway. J. Mol. Cell Biol. 2009, 1, 37–45, doi:10.1093/jmcb/mjp002.
[149]  Liu, H.L.; Zhang, Y.L.; Yang, N.; Zhang, Y.X.; Liu, X.Q.; Li, C.G.; Zhao, Y.; Wang, Y.G.; Zhang, G.G.; Yang, P.; et al. A functionalized single-walled carbon nanotube-induced autophagic cell death in human lung cells through Akt-TSC2-mTOR signaling. Cell Death Dis. 2011, 2, e159, doi:10.1038/cddis.2011.27.
[150]  Cirstea, D.; Hideshima, T.; Rodig, S.; Santo, L.; Pozzi, S.; Vallet, S.; Ikeda, H.; Perrone, G.; Gorgun, G.; Patel, K.; et al. Dual inhibition of akt/mammalian target of rapamycin pathway by nanoparticle albumin-bound-rapamycin and perifosine induces antitumor activity in multiple myeloma. Mol. Cancer Ther. 2010, 9, 963–975, doi:10.1158/1535-7163.MCT-09-0763.
[151]  Zhang, Q.; Yang, W.; Man, N.; Zheng, F.; Shen, Y.; Sun, K.; Li, Y.; Wen, L.P. Autophagy-mediated chemosensitization in cancer cells by fullerene C60 nanocrystal. Autophagy 2009, 5, 1107–1117, doi:10.4161/auto.5.8.9842.
[152]  Calzolai, L.; Franchini, F.; Gilliland, D.; Rossi, F. Protein-nanoparticle interaction: Identification of the ubiquitin—Gold nanoparticle interaction site. Nano Lett. 2010, 10, 3101–3105, doi:10.1021/nl101746v.
[153]  Ryter, S.W.; Nakahira, K.; Haspel, J.A.; Choi, A.M. Autophagy in pulmonary diseases. Annu. Rev. Physiol. 2012, 74, 377–401, doi:10.1146/annurev-physiol-020911-153348.
[154]  Seleverstov, O.; Phang, J.M.; Zabirnyk, O. Semiconductor nanocrystals in autophagy research: Methodology improvement at nanosized scale. Methods Enzymol. 2009, 452, 277–296.
[155]  Joshi, P.; Chakraborti, S.; Ramirez-Vick, J.E.; Ansari, Z.A.; Shanker, V.; Chakrabarti, P.; Singh, S.P. The anticancer activity of chloroquine-gold nanoparticles against MCF-7 breast cancer cells. Colloids Surf. B Biointerfaces 2012, 95, 195–200, doi:10.1016/j.colsurfb.2012.02.039.
[156]  Wei, P.; Zhang, L.; Lu, Y.; Man, N.; Wen, L. C60(Nd) nanoparticles enhance chemotherapeutic susceptibility of cancer cells by modulation of autophagy. Nanotechnology 2010, 21, 495101, doi:10.1088/0957-4484/21/49/495101.
[157]  Salvador-Morales, C.; Gao, W.; Ghatalia, P.; Murshed, F.; Aizu, W.; Langer, R.; Farokhzad, O.C. Multifunctional nanoparticles for prostate cancer therapy. Exp. Rev. Anticancer Ther. 2009, 9, 211–221, doi:10.1586/14737140.9.2.211.
[158]  Veale, D.; Kerr, N.; Gibson, G.J.; Harris, A.L. Characterization of epidermal growth factor receptor in primary human non-small cell lung cancer. Cancer Res. 1989, 49, 1313–1317.
[159]  Pao, W.; Miller, V.A. Epidermal growth factor receptor mutations, small-molecule kinase inhibitors, and non-small-cell lung cancer: Current knowledge and future directions. J. Clin. Oncol. 2005, 23, 2556–2568, doi:10.1200/JCO.2005.07.799.
[160]  Cappuzzo, F.; Hirsch, F.R.; Rossi, E.; Bartolini, S.; Ceresoli, G.L.; Bemis, L.; Haney, J.; Witta, S.; Danenberg, K.; Domenichini, I.; et al. Epidermal growth factor receptor gene and protein and gefitinib sensitivity in non-small-cell lung cancer. J. Natl. Cancer Inst. 2005, 97, 643–655, doi:10.1093/jnci/dji112.
[161]  Paez, J.G.; J?nne, P.A.; Lee, J.C.; Tracy, S.; Greulich, H.; Gabriel, S.; Herman, P.; Kaye, F.J.; Lindeman, N.; Boggon, T.J.; et al. EGFR mutations in lung cancer: Correlation with clinical response to gefitinib therapy. Science 2004, 304, 1497–1500, doi:10.1126/science.1099314.
[162]  Sayes, C.; Fortner, J.; Lyon, D.; Boyd, A.; Ausman, K.; Tao, Y.; Sitharaman, B.; Wilson, L.; West, J.; Colvin, V.L. The differential cytotoxicity of water soluble fullerenes. Nano Lett. 2004, 4, 1881–1887, doi:10.1021/nl0489586.
[163]  Isakovic, A.; Markovic, Z.; Todorovic-Markovic, B.; Nikolic, N.; Vranjes-Djuric, S.; Mirkovic, M.; Dramicanin, M.; Harhaji, L.; Raicevic, N.; Nikolic, Z.; et al. Distinct cytotoxic mechanisms of pristine versus hydroxylated fullerene. Toxicol. Sci. 2006, 91, 173–183, doi:10.1093/toxsci/kfj127.
[164]  Giese, A.; Bjerkvig, R.; Berens, M.E.; Westphal, M. Cost of migration: Invasion of malignant gliomas and implications for treatment. J. Clin. Oncol. 2003, 21, 1624–1636, doi:10.1200/JCO.2003.05.063.
[165]  Finn, O.J. Cancer vaccines: Between the idea and the reality. Nat. Rev. Immunol. 2003, 3, 630–641, doi:10.1038/nri1150.
[166]  Amaravadi, R.K.; Thompson, C.B. The roles of therapy-induced autophagy and necrosis in cancer treatment. Clin. Cancer Res. 2007, 13, 7271–7279, doi:10.1158/1078-0432.CCR-07-1595.

Full-Text

comments powered by Disqus

Contact Us

service@oalib.com

QQ:3279437679

WhatsApp +8615387084133

WeChat 1538708413