Isaac Scientific Publishing

Journal of Advances in Nanomaterials

Improving the Performance of Nano–Endofullerenes in Polyaniline Nanostructure–Based Biosensors by Covering Californium Colloidal Nanoparticles with Multi–Walled Carbon Nanotubes

Download PDF (1507.7 KB) PP. 1 - 28 Pub. Date: March 1, 2018

DOI: 10.22606/jan.2018.31001

Author(s)

  • Alireza Heidari*
    Faculty of Chemistry, California South University, 14731 Comet St. Irvine, CA 92604, USA

Abstract

In the current paper, Glucose (Dextrose), Fructose and Galactose–oxidase enzymes are used as stabilization medium due to its more efficiency, ability for more accurate controlling the enzyme reaction, protecting against wasting of enzyme as well as simple and easy use and exchange of enzyme medium after performing some levels of surface modification and developing Nano Endohedral Fullerenes (Endofullerenes) on Californium plate. For better connecting and stabilizing the enzyme on the medium, the prepared medium is washed by high concentration Sulfuric Acid and Nitric Acid and a large volume of deionized water and for protecting enzyme from devastating effect of Californium and prohibiting them to become inactive, surface is covered with Cystamine before stabilization. Regarding the large size of Glucose (Dextrose), Fructose and Galactose–oxidase enzymes compared to surface of medium, a connective material with amid at one end and Pyrine at the other end is used as transfer agent and for stabilizing this connection, the prepared medium is placed into dimethylformamide (DMF) solution for a couple of hours. Activity of stabilized enzyme at 460 (nm) wavelength recorded by spectroscope was depicted against time to evaluate its stability in various times. The prepared medium, which has a large amount of Glucose (Dextrose), Fructose and Galactose–oxidase enzymes, can be used as electrode in sensors. Furthermore, qualitative and quantitative measurement of food components is of great importance due to high cost of traditional methods, in addition to tendency for more accurate and sensitive detecting of these components. Glucose (Dextrose), Fructose, Galactose and Cholesterol and Cholesterol are such compounds that they frequently measure. Various methods are used to detect these food elements. However, the necessity for accurate measurement of these two compounds with high sensitivity, especially for food health issue, leads to developing biological methods, especially biosensors. Among them, biosensors based on conductive polymer nanostructures, especially Polyaniline (PANI), have been recently interested due to their unique characteristics. The current paper aims to introduce and investigate the previously performed studies about Polyaniline (PANI)–based biosensors for detecting Glucose (Dextrose), Fructose, Galactose and Cholesterol and Cholesterol. In addition, Glucose (Dextrose), Fructose and Galactose–oxidase electrochemical sensor is one of the best methods for detecting low amount of Glucose (Dextrose), Fructose and Galactose and applying Californium colloidal nanoparticles as a supplementary material in the structure of biosensor can be effective for improving its efficiency and optimum performance.

Keywords

Nano Endohedral Fullerenes (Endofullerenes), Glucose (Dextrose), Fructose and Galactose–oxidase enzymes, vapor phase precipitation, biosensor, Cholesterol, Polyaniline (PANI) nanofibers, Polyaniline (PANI) nanotubes, Californium colloidal nanoparticles, Plutonium, currentmetry

References

[1] Alireza Heidari, Christopher Brown, “Study of Composition and Morphology of Cadmium Oxide (CdO) Nanoparticles for Eliminating Cancer Cells”, Journal of Nanomedicine Research, Volume 2, Issue 5, 20 Pages, 2015.

[2] Alireza Heidari, Christopher Brown, “Study of Surface Morphological, Phytochemical and Structural Characteristics of Rhodium (III) Oxide (Rh2O3) Nanoparticles”, International Journal of Pharmacology, Phytochemistry and Ethnomedicine, Volume 1, Pages 15–19, 2015.

[3] Alireza Heidari, “An Experimental Biospectroscopic Study on Seminal Plasma in Determination of Semen Quality for Evaluation of Male Infertility”, Int J Adv Technol 7: e007, 2016.

[4] Alireza Heidari, “Extraction and Preconcentration of N–Tolyl–Sulfonyl–Phosphoramid–Saeure–Dichlorid as an Anti–Cancer Drug from Plants: A Pharmacognosy Study”, J Pharmacogn Nat Prod 2: e103, 2016.

[5] Alireza Heidari, “A Thermodynamic Study on Hydration and Dehydration of DNA and RNA?Amphiphile Complexes”, J Bioeng Biomed Sci S: 006, 2016.

[6] Alireza Heidari, “Computational Studies on Molecular Structures and Carbonyl and Ketene Groups’ Effects of Singlet and Triplet Energies of Azidoketene O=C=CH–NNN and Isocyanatoketene O=C=CH–N=C=O”, J Appl Computat Math 5: e142, 2016.

[7] Alireza Heidari, “Study of Irradiations to Enhance the Induces the Dissociation of Hydrogen Bonds between Peptide Chains and Transition from Helix Structure to Random Coil Structure Using ATR–FTIR, Raman and 1HNMR Spectroscopies”, J Biomol Res Ther 5: e146, 2016.

[8] Alireza Heidari, “Future Prospects of Point Fluorescence Spectroscopy, Fluorescence Imaging and Fluorescence Endoscopy in Photodynamic Therapy (PDT) for Cancer Cells”, J Bioanal Biomed 8: e135, 2016.

[9] Alireza Heidari, “A Bio–Spectroscopic Study of DNA Density and Color Role as Determining Factor for Absorbed Irradiation in Cancer Cells”, Adv Cancer Prev 1: e102, 2016.

[10] Alireza Heidari, “Manufacturing Process of Solar Cells Using Cadmium Oxide (CdO) and Rhodium (III) Oxide (Rh2O3) Nanoparticles”, J Biotechnol Biomater 6: e125, 2016.

[11] Alireza Heidari, “A Novel Experimental and Computational Approach to Photobiosimulation of Telomeric DNA/RNA: A Biospectroscopic and Photobiological Study”, J Res Development 4: 144, 2016.

[12] Alireza Heidari, “Biochemical and Pharmacodynamical Study of Microporous Molecularly Imprinted Polymer Selective for Vancomycin, Teicoplanin, Oritavancin, Telavancin and Dalbavancin Binding”, Biochem Physiol 5: e146, 2016.

[13] Alireza Heidari, “Anti–Cancer Effect of UV Irradiation at Presence of Cadmium Oxide (CdO) Nanoparticles on DNA of Cancer Cells: A Photodynamic Therapy Study”, Arch Cancer Res. 4: 1, 2016.

[14] Alireza Heidari, “Biospectroscopic Study on Multi–Component Reactions (MCRs) in Two A–Type and B–Type Conformations of Nucleic Acids to Determine Ligand Binding Modes, Binding Constant and Stability of Nucleic Acids in Cadmium Oxide (CdO) Nanoparticles–Nucleic Acids Complexes as Anti–Cancer Drugs”, Arch Cancer Res. 4: 2, 2016.

[15] Alireza Heidari, “Simulation of Temperature Distribution of DNA/RNA of Human Cancer Cells Using Time–Dependent Bio–Heat Equation and Nd: YAG Lasers”, Arch Cancer Res. 4: 2, 2016.

[16] Alireza Heidari, “Quantitative Structure–Activity Relationship (QSAR) Approximation for Cadmium Oxide (CdO) and Rhodium (III) Oxide (Rh2O3) Nanoparticles as Anti–Cancer Drugs for the Catalytic Formation of Proviral DNA from Viral RNA Using Multiple Linear and Non–Linear Correlation Approach”, Ann Clin Lab Res. 4: 1, 2016.

[17] Alireza Heidari, “Biomedical Study of Cancer Cells DNA Therapy Using Laser Irradiations at Presence of Intelligent Nanoparticles”, J Biomedical Sci. 5: 2, 2016.

[18] Alireza Heidari, “Measurement the Amount of Vitamin D2 (Ergocalciferol), Vitamin D3 (Cholecalciferol) and Absorbable Calcium (Ca2+), Iron (II) (Fe2+), Magnesium (Mg2+), Phosphate (PO4–) and Zinc (Zn2+) in Apricot Using High–Performance Liquid Chromatography (HPLC) and Spectroscopic Techniques”, J Biom Biostat 7: 292, 2016.

[19] Alireza Heidari, “Spectroscopy and Quantum Mechanics of the Helium Dimer (He2+), Neon Dimer (Ne2+), Argon Dimer (Ar2+), Krypton Dimer (Kr2+), Xenon Dimer (Xe2+), Radon Dimer(Rn2+) and Ununoctium Dimer (Uuo2+) Molecular Cations”, Chem Sci J 7: e112, 2016.

[20] Alireza Heidari, “Human Toxicity Photodynamic Therapy Studies on DNA/RNA Complexes as a Promising New Sensitizer for the Treatment of Malignant Tumors Using Bio–Spectroscopic Techniques”, J Drug Metab Toxicol 7: e129, 2016.

[21] Alireza Heidari, “Novel and Stable Modifications of Intelligent Cadmium Oxide (CdO) Nanoparticles as Anti–Cancer Drug in Formation of Nucleic Acids Complexes for Human Cancer Cells’ Treatment”, Biochem Pharmacol (Los Angel) 5: 207, 2016.

[22] Alireza Heidari, “A Combined Computational and QM/MM Molecular Dynamics Study on Boron Nitride Nanotubes (BNNTs), Amorphous Boron Nitride Nanotubes (a–BNNTs) and Hexagonal Boron Nitride Nanotubes (h–BNNTs) as Hydrogen Storage”, Struct Chem Crystallogr Commun 2: 1, 2016.

[23] Alireza Heidari, “Pharmaceutical and Analytical Chemistry Study of Cadmium Oxide (CdO) Nanoparticles Synthesis Methods and Properties as Anti–Cancer Drug and its Effect on Human Cancer Cells”, Pharm Anal Chem Open Access 2: 113, 2016.

[24] Alireza Heidari, “A Chemotherapeutic and Biospectroscopic Investigation of the Interaction of Double–Standard DNA/RNA–Binding Molecules with Cadmium Oxide (CdO) and Rhodium (III) Oxide (Rh2O3) Nanoparticles as Anti–Cancer Drugs for Cancer Cells’ Treatment”, Chemo Open Access 5: e129, 2016.

[25] Alireza Heidari, “Pharmacokinetics and Experimental Therapeutic Study of DNA and Other Biomolecules Using Lasers: Advantages and Applications”, J Pharmacokinet Exp Ther 1: e005, 2016.

[26] Alireza Heidari, “Determination of Ratio and Stability Constant of DNA/RNA in Human Cancer Cells and Cadmium Oxide (CdO) Nanoparticles Complexes Using Analytical Electrochemical and Spectroscopic Techniques”, Insights Anal Electrochem 2: 1, 2016.

[27] Alireza Heidari, “Discriminate between Antibacterial and Non–Antibacterial Drugs Artificial Neutral Networks of a Multilayer Perceptron (MLP) Type Using a Set of Topological Descriptors”, J Heavy Met Toxicity Dis. 1: 2, 2016.

[28] Alireza Heidari, “Combined Theoretical and Computational Study of the Belousov–Zhabotinsky Chaotic Reaction and Curtius Rearrangement for Synthesis of Mechlorethamine, Cisplatin, Streptozotocin, Cyclophosphamide, Melphalan, Busulphan and BCNU as Anti–Cancer Drugs”, Insights Med Phys. 1: 2, 2016.

[29] Alireza Heidari, “A Translational Biomedical Approach to Structural Arrangement of Amino Acids’ Complexes: A Combined Theoretical and Computational Study”, Transl Biomed. 7: 2, 2016.

[30] Alireza Heidari, “Ab Initio and Density Functional Theory (DFT) Studies of Dynamic NMR Shielding Tensors and Vibrational Frequencies of DNA/RNA and Cadmium Oxide (CdO) Nanoparticles Complexes in Human Cancer Cells”, J Nanomedine Biotherapeutic Discov 6: e144, 2016.

[31] Alireza Heidari, “Molecular Dynamics and Monte–Carlo Simulations for Replacement Sugars in Insulin Resistance, Obesity, LDL Cholesterol, Triglycerides, Metabolic Syndrome, Type 2 Diabetes and Cardiovascular Disease: A Glycobiological Study”, J Glycobiol 5: e111, 2016.

[32] Alireza Heidari, “Synthesis and Study of 5–[(Phenylsulfonyl)Amino]–1,3,4–Thiadiazole–2–Sulfonamide as Potential Anti–Pertussis Drug Using Chromatography and Spectroscopy Techniques”, Transl Med (Sunnyvale) 6: e138, 2016.

[33] Alireza Heidari, “Nitrogen, Oxygen, Phosphorus and Sulphur Heterocyclic Anti–Cancer Nano Drugs Separation in the Supercritical Fluid of Ozone (O3) Using Soave–Redlich–Kwong (SRK) and Pang–Robinson (PR) Equations”, Electronic J Biol 12: 4, 2016.

[34] Alireza Heidari, “An Analytical and Computational Infrared Spectroscopic Review of Vibrational Modes in Nucleic Acids”, Austin J Anal Pharm Chem. 3(1): 1058, 2016.

[35] Alireza Heidari, Christopher Brown, “Phase, Composition and Morphology Study and Analysis of Os–Pd/HfC Nanocomposites”, Nano Res Appl. 2: 1, 2016.

[36] Alireza Heidari, Christopher Brown, “Vibrational Spectroscopic Study of Intensities and Shifts of Symmetric Vibration Modes of Ozone Diluted by Cumene”, International Journal of Advanced Chemistry, 4 (1) 5–9, 2016.

[37] Alireza Heidari, “Study of the Role of Anti–Cancer Molecules with Different Sizes for Decreasing Corresponding Bulk Tumor Multiple Organs or Tissues”, Arch Can Res. 4: 2, 2016.

[38] Alireza Heidari, “Genomics and Proteomics Studies of Zolpidem, Necopidem, Alpidem, Saripidem, Miroprofen, Zolimidine, Olprinone and Abafungin as Anti–Tumor, Peptide Antibiotics, Antiviral and Central Nervous System (CNS) Drugs”, J Data Mining Genomics & Proteomics 7: e125, 2016.

[39] Alireza Heidari, “Pharmacogenomics and Pharmacoproteomics Studies of Phosphodiesterase–5 (PDE5) Inhibitors and Paclitaxel Albumin–Stabilized Nanoparticles as Sandwiched Anti–Cancer Nano Drugs between Two DNA/RNA Molecules of Human Cancer Cells”, J Pharmacogenomics Pharmacoproteomics 7: e153, 2016.

[40] Alireza Heidari, “Biotranslational Medical and Biospectroscopic Studies of Cadmium Oxide (CdO) Nanoparticles–DNA/RNA Straight and Cycle Chain Complexes as Potent Anti–Viral, Anti–Tumor and Anti–Microbial Drugs: A Clinical Approach”, Transl Biomed. 7: 2, 2016.

[41] Alireza Heidari, “A Comparative Study on Simultaneous Determination and Separation of Adsorbed Cadmium Oxide (CdO) Nanoparticles on DNA/RNA of Human Cancer Cells Using Biospectroscopic Techniques and Dielectrophoresis (DEP) Method”, Arch Can Res. 4: 2, 2016.

[42] Alireza Heidari, “Cheminformatics and System Chemistry of Cisplatin, Carboplatin, Nedaplatin, Oxaliplatin, Heptaplatin and Lobaplatin as Anti–Cancer Nano Drugs: A Combined Computational and Experimental Study”, J Inform Data Min 1: 3, 2016.

[43] Alireza Heidari, “Linear and Non–Linear Quantitative Structure–Anti–Cancer–Activity Relationship (QSACAR) Study of Hydrous Ruthenium (IV) Oxide (RuO2) Nanoparticles as Non–Nucleoside Reverse Transcriptase Inhibitors (NNRTIs) and Anti–Cancer Nano Drugs”, J Integr Oncol 5: e110, 2016.

[44] Alireza Heidari, “Synthesis, Characterization and Biospectroscopic Studies of Cadmium Oxide (CdO) Nanoparticles–Nucleic Acids Complexes Absence of Soluble Polymer as a Protective Agent Using Nucleic Acids Condensation and Solution Reduction Method”, J Nanosci Curr Res 1: e101, 2016.

[45] Alireza Heidari, “Coplanarity and Collinearity of 4’–Dinonyl–2,2’–Bithiazole in One Domain of Bleomycin and Pingyangmycin to be Responsible for Binding of Cadmium Oxide (CdO) Nanoparticles to DNA/RNA Bidentate Ligands as Anti–Tumor Nano Drug”, Int J Drug Dev & Res 8: 007–008, 2016.

[46] Alireza Heidari, “A Pharmacovigilance Study on Linear and Non–Linear Quantitative Structure (Chromatographic) Retention Relationships (QSRR) Models for the Prediction of Retention Time of Anti–Cancer Nano Drugs under Synchrotron Radiations”, J Pharmacovigil 4: e161, 2016.

[47] Alireza Heidari, “Nanotechnology in Preparation of Semipermeable Polymers”, J Adv Chem Eng 6: 157, 2016.

[48] Alireza Heidari, “A Gastrointestinal Study on Linear and Non–Linear Quantitative Structure (Chromatographic) Retention Relationships (QSRR) Models for Analysis 5–Aminosalicylates Nano Particles as Digestive System Nano Drugs under Synchrotron Radiations”, J Gastrointest Dig Syst 6: e119, 2016.

[49] Alireza Heidari, “DNA/RNA Fragmentation and Cytolysis in Human Cancer Cells Treated with Diphthamide Nano Particles Derivatives”, Biomedical Data Mining 5: e102, 2016.

[50] Alireza Heidari, “A Successful Strategy for the Prediction of Solubility in the Construction of Quantitative Structure–Activity Relationship (QSAR) and Quantitative Structure–Property Relationship (QSPR) under Synchrotron Radiations Using Genetic Function Approximation (GFA) Algorithm”, J Mol Biol Biotechnol 1: 1, 2016.

[51] Alireza Heidari, “Computational Study on Molecular Structures of C20, C60, C240, C540, C960, C2160 and C3840 Fullerene Nano Molecules under Synchrotron Radiations Using Fuzzy Logic”, J Material Sci Eng 5: 282, 2016.

[52] Alireza Heidari, “Graph Theoretical Analysis of Zigzag Polyhexamethylene Biguanide, Polyhexamethylene Adipamide, Polyhexamethylene Biguanide Gauze and Polyhexamethylene Biguanide Hydrochloride (PHMB) Boron Nitride Nanotubes (BNNTs), Amorphous Boron Nitride Nanotubes (a–BNNTs) and Hexagonal Boron Nitride Nanotubes (h–BNNTs)”, J Appl Computat Math 5: e143, 2016.

[53] Alireza Heidari, “The Impact of High Resolution Imaging on Diagnosis”, Int J Clin Med Imaging 3: 1000e101, 2016.

[54] Alireza Heidari, “A Comparative Study of Conformational Behavior of Isotretinoin (13–Cis Retinoic Acid) and Tretinoin (All–Trans Retinoic Acid (ATRA)) Nano Particles as Anti–Cancer Nano Drugs under Synchrotron Radiations Using Hartree–Fock (HF) and Density Functional Theory (DFT) Methods”, Insights in Biomed 1: 2, 2016.

[55] Alireza Heidari, “Advances in Logic, Operations and Computational Mathematics”, J Appl Computat Math 5: 5, 2016.

[56] Alireza Heidari, “Mathematical Equations in Predicting Physical Behavior”, J Appl Computat Math 5: 5, 2016.

[57] Alireza Heidari, “Chemotherapy a Last Resort for Cancer Treatment”, Chemo Open Access 5: 4, 2016.

[58] Alireza Heidari, “Separation and Pre–Concentration of Metal Cations–DNA/RNA Chelates Using Molecular Beam Mass Spectrometry with Tunable Vacuum Ultraviolet (VUV) Synchrotron Radiation and Various Analytical Methods”, Mass Spectrom Purif Tech 2: e101, 2016.

[59] Alireza Heidari, “Yoctosecond Quantitative Structure–Activity Relationship (QSAR) and Quantitative Structure–Property Relationship (QSPR) under Synchrotron Radiations Studies for Prediction of Solubility of Anti–Cancer Nano Drugs in Aqueous Solutions Using Genetic Function Approximation (GFA) Algorithm”, Insight Pharm Res. 1: 1, 2016.

[60] Alireza Heidari, “Cancer Risk Prediction and Assessment in Human Cells under Synchrotron Radiations Using Quantitative Structure Activity Relationship (QSAR) and Quantitative Structure Properties Relationship (QSPR) Studies”, Int J Clin Med Imaging 3: 516, 2016.

[61] T. Bastogne, Quality-by-design of nanopharmaceuticals – a state of the art, Nanomedicine: Nanotechnology, Biology and Medicine, 2017, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2017.05.014.

[62] ?eljka Vani?, Nata?a ?kalko-Basnet, Nanopharmaceuticals for improved topical vaginal therapy: Can they deliver?, European Journal of Pharmaceutical Sciences, Volume 50, Issue 1, 2013, Pages 29-41, ISSN 0928-0987, http://dx.doi.org/10.1016/j.ejps.2013.04.035.

[63] German A. Islan, Marcela Durán, Maximiliano L. Cacicedo, Gerson Nakazato, Renata K.T. Kobayashi, Diego S.T. Martinez, Guillermo R. Castro, Nelson Durán, Nanopharmaceuticals as a solution to neglected diseases: Is it possible?, Acta Tropica, Volume 170, 2017, Pages 16-42, ISSN 0001-706X, http://dx.doi.org/10.1016/ j.actatropica.2017.02.019.

[64] Willie E. Bawarski, Elena Chidlowsky, Dhruba J. Bharali, Shaker A. Mousa, Emerging nanopharmaceuticals, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 4, Issue 4, 2008, Pages 273-282, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2008.06.002.

[65] Michael A.W. Eaton, How do we develop nanopharmaceuticals under open innovation?, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 7, Issue 4, 2011, Pages 371-375, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2011.05.015.

[66] Kunn Hadinoto, Yue Yang, Continuous and sustainable granulation of nanopharmaceuticals by spray coagulation encapsulation in alginate, International Journal of Pharmaceutics, Volume 473, Issue 1, 2014, Pages 644-652, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2014.07.042.

[67] Sonke Svenson, Marc Wolfgang, Jungyeon Hwang, John Ryan, Scott Eliasof, Preclinical to clinical development of the novel camptothecin nanopharmaceutical CRLX101, Journal of Controlled Release, Volume 153, Issue 1, 2011, Pages 49-55, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2011.03.007.

[68] Alejandro Sosnik, Reversal of multidrug resistance by the inhibition of ATP-binding cassette pumps employing “Generally Recognized As Safe” (GRAS) nanopharmaceuticals: A review, Advanced Drug Delivery Reviews, Volume 65, Issue 13, Pages 1828-1851, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2013.09.002.

[69] Jelena Filipovi?-Gr?i?, Ale? Mrhar, Hans Junginger, Thematic Issue on Emerging nanopharmaceuticals for non-parenteral application routes, European Journal of Pharmaceutical Sciences, Volume 50, Issue 1, 2013, Page 1, ISSN 0928-0987, http://dx.doi.org/10.1016/j.ejps.2013.05.025.

[70] Hong Yu, Kunn Hadinoto, Mitigating the adverse effect of spray drying on the supersaturation generation capability of amorphous nanopharmaceutical powders, Powder Technology, Volume 277, 2015, Pages 97-104, ISSN 0032-5910, http://dx.doi.org/10.1016/j.powtec.2015.02.059.

[71] S. Moein Moghimi, Z. Shadi Farhangrazi, Nanomedicine and the complement paradigm, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 9, Issue 4, 2013, Pages 458-460, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2013.02.011.

[72] S. Eliasof, P.S. Ng, P. Lim Soo, J. Podobinski, R.I. Case, P. Shum, J.G. Martinez, S.R. Kabir, D. Lazarus, S. Svenson, 425 Significantly enhanced therapeutic profile of docetaxel in novel nanopharmaceutical CRLX288, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Page 135, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72132-7.

[73] Concepción Domingo, Javier Saurina, An overview of the analytical characterization of nanostructured drug delivery systems: Towards green and sustainable pharmaceuticals: A review, Analytica Chimica Acta, Volume 744, 2012, Pages 8-22, ISSN 0003-2670, http://dx.doi.org/10.1016/j.aca.2012.07.010.

[74] Asmita Samadder, Suresh K. Abraham, Anisur Rahman Khuda-Bukhsh, Nanopharmaceutical approach using pelargonidin towards enhancement of efficacy for prevention of alloxan-induced DNA damage in L6 cells via activation of PARP and p53, Environmental Toxicology and Pharmacology, Volume 43, 2016, Pages 27-37, ISSN 1382-6689, http://dx.doi.org/10.1016/j.etap.2016.02.010.

[75] Y. Yen, T. Synold, G.J. Weiss, T. Schluep, J. Ryan, 423 Phase 1 dose escalation, safety and pharmacokinetic study of IT-101 (CRLX101), a novel nanopharmaceutical containing camptothecin, in advanced solid tumor cancer patients, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Pages 134-135, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72130-3.

[76] Intan D.M. Azmi, Peter P. Wibroe, Lin-Ping Wu, Ali I. Kazem, Heinz Amenitsch, Seyed M. Moghimi, Anan Yaghmur, A structurally diverse library of safe-by-design citrem-phospholipid lamellar and non-lamellar liquid crystalline nano-assemblies, Journal of Controlled Release, Volume 239, 2016, Pages 1-9, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2016.08.011.

[77] Jun Li, Yujue Wang, Ruijing Liang, Xiangjie An, Ke Wang, Guanxin Shen, Yating Tu, Jintao Zhu, Juan Tao, Recent advances in targeted nanoparticles drug delivery to melanoma, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 11, Issue 3, 2015, Pages 769-794, ISSN 1549-9634, http://dx.doi.org/10.1016/ j.nano.2014.11.006.

[78] Jinhua Liu, Yongxing Zhao, Qianqian Guo, Zhao Wang, Huiyuan Wang, Yongxin Yang, Yongzhuo Huang, TAT-modified nanosilver for combating multidrug-resistant cancer, Biomaterials, Volume 33, Issue 26, 2012, Pages 6155-6161, ISSN 0142-9612, http://dx.doi.org/10.1016/j.biomaterials.2012.05.035.

[79] Cristina Gabellieri, Heico Frima, Nanomedicine in the European Commission policy for nanotechnology, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 7, Issue 5, 2011, Pages 519-520, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2011.07.003.

[80] Robert M. Frederickson, SM Moghimi, E Wagner, Seppo Yla-Herttuala, Call for papers: Nanoparticle Development and Applications in Cellular and Molecular Therapies, Molecular Therapy, Volume 24, Issue 8, 2016, Pages 1334-1335, ISSN 1525-0016, http://dx.doi.org/10.1038/mt.2016.164.

[81] Mehrdad Namdari, Ali Eatemadi, Maryam Soleimaninejad, Aiyelabegan T. Hammed, A brief review on the application of nanoparticle enclosed herbal medicine for the treatment of infective endocarditis, Biomedicine & Pharmacotherapy, Volume 87, 2017, Pages 321-331, ISSN 0753-3322, http://dx.doi.org/10.1016/ j.biopha.2016.12.099.

[82] Tie Yi Kiew, Wean Sin Cheow, Kunn Hadinoto, Preserving the supersaturation generation capability of amorphous drug-polysaccharide nanoparticle complex after freeze drying, International Journal of Pharmaceutics, Volume 484, Issue 1, 2015, Pages 115-123, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2015.02.057.

[83] S. Moein Moghimi, Peter P. Wibroe, Shen Y. Helvig, Z. Shadi Farhangrazi, A. Christy Hunter, Genomic perspectives in inter-individual adverse responses following nanomedicine administration: The way forward, Advanced Drug Delivery Reviews, Volume 64, Issue 13, 2012, Pages 1385-1393, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2012.05.010.

[84] Pilar Rivera Gil, Dominik Hühn, Loretta L. del Mercato, Daniel Sasse, Wolfgang J. Parak, Nanopharmacy: Inorganic nanoscale devices as vectors and active compounds, Pharmacological Research, Volume 62, Issue 2, 2010, Pages 115-125, ISSN 1043-6618, http://dx.doi.org/10.1016/j.phrs.2010.01.009.

[85] Beverly A. Rzigalinski, Jeannine S. Strobl, Cadmium-containing nanoparticles: Perspectives on pharmacology and toxicology of quantum dots, Toxicology and Applied Pharmacology, Volume 238, Issue 3, 2009, Pages 280-288, ISSN 0041-008X, http://dx.doi.org/10.1016/j.taap.2009.04.010.

[86] Valerie E. Fako, Darin Y. Furgeson, Zebrafish as a correlative and predictive model for assessing biomaterial nanotoxicity, Advanced Drug Delivery Reviews, Volume 61, Issue 6, 2009, Pages 478-486, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2009.03.008.

[87] Vanessa Sainz, Jo?o Conniot, Ana I. Matos, Carina Peres, Eva Zupanǒiǒ, Liane Moura, Liana C. Silva, Helena F. Florindo, Rogério S. Gaspar, Regulatory aspects on nanomedicines, Biochemical and Biophysical Research Communications, Volume 468, Issue 3, 2015, Pages 504-510, ISSN 0006-291X, http://dx.doi.org/10.1016/ j.bbrc.2015.08.023.

[88] Ruth Duncan, María J. Vicent, Do HPMA copolymer conjugates have a future as clinically useful nanomedicines? A critical overview of current status and future opportunities, Advanced Drug Delivery Reviews, Volume 62, Issue 2, 2010, Pages 272-282, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2009.12.005.

[89] Xing Zhou, Ling Che, Yanling Wei, Yin Dou, Sha Chen, Hongmei He, Hao Gong, Xiaohui Li, Jianxiang Zhang, Facile route to versatile nanoplatforms for drug delivery by one-pot self-assembly, Acta Biomaterialia, Volume 10, Issue 6, 2014, Pages 2630-2642, ISSN 1742-7061, http://dx.doi.org/10.1016/j.actbio.2014.01.024.

[90] Peter P. Wibroe, Davoud Ahmadvand, Mohammad Ali Oghabian, Anan Yaghmur, S. Moein Moghimi, An integrated assessment of morphology, size, and complement activation of the PEGylated liposomal doxorubicin products Doxil?, Caelyx?, DOXOrubicin, and SinaDoxosome, Journal of Controlled Release, Volume 221, 2016, Pages 1-8, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2015.11.021.

[91] Minh-Hiep Nguyen, Hong Yu, Bingxue Dong, Kunn Hadinoto, A supersaturating delivery system of silibinin exhibiting high payload achieved by amorphous nano-complexation with chitosan, European Journal of Pharmaceutical Sciences, Volume 89, 2016, Pages 163-171, ISSN 0928-0987, http://dx.doi.org/10.1016/ j.ejps.2016.04.036.

[92] Special Issue Title Page, Biotechnology Advances, Volume 32, Issue 4, 2014, Page iii, ISSN 0734-9750, http://dx.doi.org/10.1016/S0734-9750(14)00084-6.

[93] Mariana Beija, Robert Salvayre, Nancy Lauth-de Viguerie, Jean-Daniel Marty, Colloidal systems for drug delivery: from design to therapy, Trends in Biotechnology, Volume 30, Issue 9, 2012, Pages 485-496, ISSN 0167-7799, http://dx.doi.org/10.1016/j.tibtech.2012.04.008.

[94] A. Vaishali, K. Madhu Varma, P. Arun Bhupathi, T. Sreenivasa Bharath, M.V. Ramesh, P. Venkata Karteek Varma, In vitro evaluation of antimicrobial efficacy of 2% chlorhexidine loaded electrospun nanofibers, Journal of Pierre Fauchard Academy (India Section), 2017, ISSN 0970-2199, http://dx.doi.org/10.1016/j.jpfa.2017.01.006.

[95] Raj Bawa, NanoBiotech 2008: Exploring global advances in nanomedicine, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 5, Issue 1, 2009, Pages 5-7, ISSN 1549-9634, http://dx.doi.org/10.1016/ j.nano.2009.01.004.

[96] Carlotta Marianecci, Stefania Petralito, Federica Rinaldi, Patrizia N. Hanieh, Maria Carafa, Some recent advances on liposomal and niosomal vesicular carriers, Journal of Drug Delivery Science and Technology, Volume 32, 2016, Pages 256-269, ISSN 1773-2247, http://dx.doi.org/10.1016/j.jddst.2015.10.008.

[97] Sharvil Patil, Khushbu Chaudhari, Ravindra Kamble, Electrospray technique for cocrystallization of phytomolecules, Journal of King Saud University - Science, 2017, ISSN 1018-3647, http://dx.doi.org/10.1016/ j.jksus.2017.04.001.

[98] Nuno A. Fonseca, Ana C. Gregório, ?ngela Valério-Fernandes, Sérgio Sim?es, Jo?o N. Moreira, Bridging cancer biology and the patients' needs with nanotechnology-based approaches, Cancer Treatment Reviews, Volume 40, Issue 5, 2014, Pages 626-635, ISSN 0305-7372, http://dx.doi.org/10.1016/j.ctrv.2014.02.002.

[99] Deepa Bedi, Tiziana Musacchio, Olusegun A. Fagbohun, James W. Gillespie, Patricia Deinnocentes, R. Curtis Bird, Lonnie Bookbinder, Vladimir P. Torchilin, Valery A. Petrenko, Delivery of siRNA into breast cancer cells via phage fusion protein-targeted liposomes, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 7, Issue 3, 2011, Pages 315-323, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2010.10.004.

[100] Fabiana Canal, Joaquin Sanchis, María J Vicent, Polymer–drug conjugates as nano-sized medicines, Current Opinion in Biotechnology, Volume 22, Issue 6, 2011, Pages 894-900, ISSN 0958-1669, http://dx.doi.org/10.1016/ j.copbio.2011.06.003.

[101] Helmut M. Hügel, Neale Jackson, Danshen diversity defeating dementia, Bioorganic & Medicinal Chemistry Letters, Volume 24, Issue 3, 2014, Pages 708-716, ISSN 0960-894X, http://dx.doi.org/10.1016/j.bmcl.2013.12.042.

[102] Special Issue title page, European Journal of Pharmaceutics and Biopharmaceutics, Volume 79, Issue 1, 2011, Page v, ISSN 0939-6411, http://dx.doi.org/10.1016/S0939-6411(11)00237-2.

[103] Laurie Donaldson, Designer nanoparticles to treat blood cancer, Materials Today, Volume 15, Issue 7, 2012, Page 298, ISSN 1369-7021, http://dx.doi.org/10.1016/S1369-7021(12)70128-1.

[104] Graphical Abstracts, Journal of Fluorine Chemistry, Volume 198, 2017, Pages v-viii, ISSN 0022-1139, http://dx.doi.org/10.1016/S0022-1139(17)30214-2.

[105] Rajendran J.C. Bose, Soo-Hong Lee, Hansoo Park, Biofunctionalized nanoparticles: an emerging drug delivery platform for various disease treatments, Drug Discovery Today, Volume 21, Issue 8, 2016, Pages 1303-1312, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2016.06.005.

[106] Arnaldur Hall, Ulrich L?chelt, Jiri Bartek, Ernst Wagner, Seyed Moein Moghimi, Polyplex Evolution: Understanding Biology, Optimizing Performance, Molecular Therapy, Volume 25, Issue 7, 2017, Pages 1476-1490, ISSN 1525-0016, http://dx.doi.org/10.1016/j.ymthe.2017.01.024.

[107] Gert Storm, Preface, European Journal of Pharmaceutical Sciences, Volume 45, Issue 4, 2012, Page 387, ISSN 0928-0987, http://dx.doi.org/10.1016/j.ejps.2011.11.001.

[108] Table of Contents, Acta Pharmaceutica Sinica B, Volume 7, Issue 3, 2017, Pages iii-vii, ISSN 2211-3835, http://dx.doi.org/10.1016/S2211-3835(17)30142-9. 1

[109] Lisa C. du Toit, Viness Pillay, Yahya E. Choonara, Nano-microbicides: Challenges in drug delivery, patient ethics and intellectual property in the war against HIV/AIDS, Advanced Drug Delivery Reviews, Volume 62, Issue 4, 2010, Pages 532-546, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2009.11.022.

[110] Brajesh Kumar, Yolanda Angulo, Kumari Smita, Luis Cumbal, Alexis Debut, Capuli cherry-mediated green synthesis of silver nanoparticles under white solar and blue LED light, Particuology, Volume 24, 2016, Pages 123-128, ISSN 1674-2001, http://dx.doi.org/10.1016/j.partic.2015.05.005.

[111] Mehdi Rajabi, Thangirala Sudha, Noureldien H.E. Darwish, Paul J. Davis, Shaker A. Mousa, Synthesis of MR-49, a deiodinated analog of tetraiodothyroacetic acid (tetrac), as a novel pro-angiogenesis modulator, Bioorganic & Medicinal Chemistry Letters, Volume 26, Issue 16, 2016, Pages 4112-4116, ISSN 0960-894X, http://dx.doi.org/ 10.1016/j.bmcl.2016.06.064.

[112] Alina J. Andersen, Peter P. Wibroe, S. Moein Moghimi, Perspectives on carbon nanotube-mediated adverse immune effects, Advanced Drug Delivery Reviews, Volume 64, Issue 15, 2012, Pages 1700-1705, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2012.05.005.

[113] Subject Index Volume 153, Journal of Controlled Release, Volume 153, Issue 3, 2011, Pages e8-e9, ISSN 0168-3659, http://dx.doi.org/10.1016/S0168-3659(11)00517-7.

[114] Alexander Kabanov, Tatiana Bronich, Eighth International Nanomedicine and Drug Delivery Symposium (NanoDDS'10), Journal of Controlled Release, Volume 153, Issue 1, 2011, Page 1, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2011.06.002.

[115] Zsombor K. Nagy, Attila Balogh, Balázs Démuth, Hajnalka Pataki, Tamás Vigh, Bence Szabó, Kolos Molnár, Bence T. Schmidt, Péter Horák, Gy?rgy Marosi, Geert Verreck, Ivo Van Assche, Marcus E. Brewster, High speed electrospinning for scaled-up production of amorphous solid dispersion of itraconazole, International Journal of Pharmaceutics, Volume 480, Issue 1, 2015, Pages 137-142, ISSN 0378-5173, http://dx.doi.org/10.1016/ j.ijpharm.2015.01.025.

[116] Sharon M. Nickols-Richardson, Nanotechnology: Implications for Food and Nutrition Professionals, Journal of the American Dietetic Association, Volume 107, Issue 9, 2007, Pages 1494-1497, ISSN 0002-8223, http://dx.doi.org/10.1016/j.jada.2007.06.016.

[117] Rogério Gaspar, Ruth Duncan, Polymeric carriers: Preclinical safety and the regulatory implications for design and development of polymer therapeutics, Advanced Drug Delivery Reviews, Volume 61, Issue 13, 2009, Pages 1220-1231, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2009.06.003.

[118] Graphical Abstracts Contents Listing, Journal of Controlled Release, Volume 153, Issue 1, 2011, Pages e1-e4, ISSN 0168-3659, http://dx.doi.org/10.1016/S0168-3659(11)00428-7.

[119] Athanasios B. Bourlinos, Vasilios Georgakilas, Aristides Bakandritsos, Antonios Kouloumpis, Dimitrios Gournis, Radek Zboril, Aqueous-dispersible fullerol-carbon nanotube hybrids, Materials Letters, Volume 82, 2012, Pages 48-50, ISSN 0167-577X, http://dx.doi.org/10.1016/j.matlet.2012.05.026.

[120] Recommended Articles, Journal of Acupuncture and Meridian Studies, Volume 9, Issue 6, 2016, Pages 345-348, ISSN 2005-2901, http://dx.doi.org/10.1016/j.jams.2016.12.001.

[121] Sonke Svenson, Clinical translation of nanomedicines, Current Opinion in Solid State and Materials Science, Volume 16, Issue 6, 2012, Pages 287-294, ISSN 1359-0286, http://dx.doi.org/10.1016/j.cossms.2012.10.001.

[122] Johannes Sitterberg, Aybike ?zcetin, Carsten Ehrhardt, Udo Bakowsky, Utilising atomic force microscopy for the characterisation of nanoscale drug delivery systems, European Journal of Pharmaceutics and Biopharmaceutics, Volume 74, Issue 1, 2010, Pages 2-13, ISSN 0939-6411, http://dx.doi.org/10.1016/ j.ejpb.2009.09.005.

[123] Mark Telford, Cancer centers founded, Materials Today, Volume 8, Issue 12, 2005, Page 19, ISSN 1369-7021, http://dx.doi.org/10.1016/S1369-7021(05)71277-3.

[124] Mona Alibolandi, Fatemeh Sadeghi, Khalil Abnous, Fatemeh Atyabi, Mohammad Ramezani, Farzin Hadizadeh, The chemotherapeutic potential of doxorubicin-loaded PEG-b-PLGA nanopolymersomes in mouse breast cancer model, European Journal of Pharmaceutics and Biopharmaceutics, Volume 94, 2015, Pages 521-531, ISSN 0939-6411, http://dx.doi.org/10.1016/j.ejpb.2015.07.005.

[125] Alexandre Bridoux, Huadong Cui, Evgeny Dyskin, Murat Yalcin, Shaker A. Mousa, Semisynthesis and pharmacological activities of Tetrac analogs: Angiogenesis modulators, Bioorganic & Medicinal Chemistry Letters, Volume 19, Issue 12, 2009, Pages 3259-3263, ISSN 0960-894X, http://dx.doi.org/10.1016/j.bmcl.2009.04.094.

[126] F.E. Stuurman, E.E. Voest, A. Awada, J.H.M. Schellens, P.O. Witteveen, T. Bergeland, P.A. Hals, A. Hendlisz, 426 Phase I study of oral CP-4126, a gemcitabine analog, in patients with advanced solid tumours, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Page 135, ISSN 1359-6349, http://dx.doi.org/10.1016/ S1359-6349(10)72133-9.

[127] E. Kondo, 424 Development of novel cancer cell-selective cell-penetrating peptides for the advanced peptide-based drug delivery system, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Page 135, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72131-5.

[128] Recommended Articles, Journal of Acupuncture and Meridian Studies, Volume 9, Issue 5, 2016, Pages 281-284, ISSN 2005-2901, http://dx.doi.org/10.1016/j.jams.2016.10.002.

[129] Anil B. Jindal, Sagar S. Bachhav, Padma V. Devarajan, hybrid nano drug delivery system (IHN-DDS) of antiretroviral drug for simultaneous targeting to multiple viral reservoirs: An proof of concept, International Journal of Pharmaceutics, Volume 521, Issue 1, 2017, Pages 196-203, ISSN 0378-5173, http://dx.doi.org/10.1016/ j.ijpharm.2017.02.024.

[130] Natalya Rapoport, Physical stimuli-responsive polymeric micelles for anti-cancer drug delivery, Progress in Polymer Science, Volume 32, Issue 8, 2007, Pages 962-990, ISSN 0079-6700, http://dx.doi.org/10.1016/ j.progpolymsci.2007.05.009.

[131] Luis ángel Fernández, Serge Muyldermans, Recent developments in engineering and delivery of protein and antibody therapeutics, Current Opinion in Biotechnology, Volume 22, Issue 6, 2011, Pages 839-842, ISSN 0958-1669, http://dx.doi.org/10.1016/j.copbio.2011.08.001.

[132] Natassa Pippa, Aristides Dokoumetzidis, Costas Demetzos, Panos Macheras, On the ubiquitous presence of fractals and fractal concepts in pharmaceutical sciences: A review, International Journal of Pharmaceutics, Volume 456, Issue 2, 2013, Pages 340-352, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2013.08.087.

[133] M. Verreault, D. Strutt, D. Masin, M. Anantha, D. Waterhouse, D.T. Yapp, M.B. Bally, Irinophore C?, a lipid-based nanoparticulate formulation of irinotecan, is more effective than free irinotecan when used to treat an orthotopic glioblastoma model, Journal of Controlled Release, Volume 158, Issue 1, 2012, Pages 34-43, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2011.09.095.

[134] Parichehr Hassanzadeh, Fatemeh Atyabi, Rassoul Dinarvand, Application of modelling and nanotechnology-based approaches: The emergence of breakthroughs in theranostics of central nervous system disorders, Life Sciences, Volume 182, 2017, Pages 93-103, ISSN 0024-3205, http://dx.doi.org/10.1016/j.lfs.2017.06.001.

[135] Senthilkumar Sivanesan, Aaron Tan, Rebecca Jeyaraj, James Lam, Monica Gole, Antonio Hardan, Keyoumars Ashkan, Jayakumar Rajadas, Pharmaceuticals and Stem Cells in Autism Spectrum Disorders: Wishful Thinking?, World Neurosurgery, Volume 98, 2017, Pages 659-672, ISSN 1878-8750, http://dx.doi.org/10.1016/ j.wneu.2016.09.100.

[136] R. Phillips, H. Makeen, N. Periasamy, P. Loadman, S. Smye, B. Sleeman, P. Jones, C. Evans, C. Twelves, 427 The development and evaluation of an experimental model for assessing convective fluid flow through multicell layers, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Pages 135-136, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72134-0.

[137] Gamze Varan, Cem Varan, Nazl? Erdo?ar, A. Atilla H?ncal, Erem Bilensoy, Amphiphilic cyclodextrin nanoparticles, International Journal of Pharmaceutics, 2017, ISSN 0378-5173, http://dx.doi.org/10.1016/ j.ijpharm.2017.06.010.

[138] Seyed Moein Moghimi, Zahra Shadi Farhangrazi, Just so stories: The random acts of anti-cancer nanomedicine performance, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 10, Issue 8, 2014, Pages 1661-1666, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2014.04.011.

[139] J.C. Soria, C.A. Gomez-Roca, J.A. Ware, A.A. Adjei, R.K. Brachmann, H.J.M. Groen, 421 A Phase Ib study to evaluate the pan-PI3K inhibitor GDC-0941 with paclitaxel and carboplatin with and without bevacizumab in non-small cell lung cancer patients, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Page 134, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72128-5.

[140] J. McMurray, J. Klostergaard, E.J. Auzenne, W.S.L. Liao, Z. Lu, P.K. Mandal, R. Ramesh, M. Shanker, A.W. Scott, 422 Targeting the SH2 domain of Stat3 with phosphopeptide mimetic prodrugs leads to tumor growth inhibition and down-regulation of phosphoTyr705 Stat3 and angiogenic pathways, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Page 134, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72129-7.

[141] E. Sans-Serramitjana, E. Fusté, B. Martínez-Garriga, A. Merlos, M. Pastor, J.L. Pedraz, A. Esquisabel, D. Bachiller, T. Vinuesa, M. Vi?as, Killing effect of nanoencapsulated colistin sulfate on from cystic fibrosis patients, Journal of Cystic Fibrosis, Volume 15, Issue 5, 2016, Pages 611-618, ISSN 1569-1993, http://dx.doi.org/10.1016/ j.jcf.2015.12.005.

[142] Lucas A. Rigo, Cristiane S. Carvalho-Wodarz, Adriana R. Pohlmann, Silvia S. Guterres, Nicole Schneider-Daum, Claus-Michael Lehr, Ruy C.R. Beck, Nanoencapsulation of a glucocorticoid improves barrier function and anti-inflammatory effect on monolayers of pulmonary epithelial cell lines, European Journal of Pharmaceutics and Biopharmaceutics, Volume 119, 2017, Pages 1-10, ISSN 0939-6411, http://dx.doi.org/10.1016/j.ejpb.2017.05.006.

[143] Mona Alibolandi, Seyed Mohammad Taghdisi, Pouria Ramezani, Fazileh Hosseini Shamili, Sara Amel Farzad, Khalil Abnous, Mohammad Ramezani, Smart AS1411-aptamer conjugated pegylated PAMAM dendrimer for the superior delivery of camptothecin to colon adenocarcinoma and , International Journal of Pharmaceutics, Volume 519, Issue 1, 2017, Pages 352-364, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2017.01.044.

[144] Alexandre Bridoux, Huadong Cui, Evgeny Dyskin, Andreea-Ruxandra Schmitzer, Murat Yalcin, Shaker A. Mousa, Semisynthesis and pharmacological activities of thyroxine analogs: Development of new angiogenesis modulators, Bioorganic & Medicinal Chemistry Letters, Volume 20, Issue 11, 2010, Pages 3394-3398, ISSN 0960-894X, http://dx.doi.org/10.1016/j.bmcl.2010.04.011.

[145] Krzysztof Tutaj, Radoslaw Szlazak, Katarzyna Szalapata, Joanna Starzyk, Rafal Luchowski, Wojciech Grudzinski, Monika Osinska-Jaroszuk, Anna Jarosz-Wilkolazka, Agnieszka Szuster-Ciesielska, Wieslaw I. Gruszecki, Amphotericin B-silver hybrid nanoparticles: synthesis, properties and antifungal activity, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 12, Issue 4, 2016, Pages 1095-1103, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2015.12.378.

[146] Palaniselvam Kuppusamy, Mashitah M. Yusoff, Gaanty Pragas Maniam, Natanamurugaraj Govindan, A case study – Regulation and functional mechanisms of cancer cells and control its activity using plants and their derivatives, Journal of Pharmacy Research, Volume 6, Issue 8, 2013, Pages 884-892, ISSN 0974-6943, http://dx.doi.org/10.1016/j.jopr.2013.08.002.

[147] Donald A. Tomalia, International report on nanomedicine in the U.S.A., Nanomedicine: Nanotechnology, Biology and Medicine, Volume 2, Issue 4, 2006, Page 299, ISSN 1549-9634, http://dx.doi.org/10.1016/ j.nano.2006.10.098.

[148] Anupa R. Menjoge, Rangaramanujam M. Kannan, Donald A. Tomalia, Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications, Drug Discovery Today, Volume 15, Issue 5, 2010, Pages 171-185, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2010.01.009.

[149] Karina R. Vega-Villa, Jody K. Takemoto, Jaime A. Yá?ez, Connie M. Remsberg, M. Laird Forrest, Neal M. Davies, Clinical toxicities of nanocarrier systems, Advanced Drug Delivery Reviews, Volume 60, Issue 8, 2008, Pages 929-938, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2007.11.007.

[150] Shikha Gaur, Yafan Wang, Leo Kretzner, Linling Chen, Terence Yen, Xiwei Wu, Yate-Ching Yuan, Mark Davis, Yun Yen, Pharmacodynamic and pharmacogenomic study of the nanoparticle conjugate of camptothecin CRLX101 for the treatment of cancer, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 10, Issue 7, 2014, Pages 1477-1486, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2014.04.003.

[151] Rainer Tietze, Jan Zaloga, Harald Unterweger, Stefan Lyer, Ralf P. Friedrich, Christina Janko, Marina P?ttler, Stephan Dürr, Christoph Alexiou, Magnetic nanoparticle-based drug delivery for cancer therapy, Biochemical and Biophysical Research Communications, Volume 468, Issue 3, 2015, Pages 463-470, ISSN 0006-291X, http://dx.doi.org/10.1016/j.bbrc.2015.08.022.

[152] Alex Schwengber, Héctor J. Prado, Darío A. Zilli, Pablo R. Bonelli, Ana L. Cukierman, Carbon nanotubes buckypapers for potential transdermal drug delivery, Materials Science and Engineering: C, Volume 57, 2015, Pages 7-13, ISSN 0928-4931, http://dx.doi.org/10.1016/j.msec.2015.07.030.

[153] Piya Adhikari, Paulami Pal, Anup Kr. Das, Subhabrata Ray, Arpita Bhattacharjee, Bhaskar Mazumder, NANO LIPID-DRUG CONJUGATE: AN INTEGRATED REVIEW, International Journal of Pharmaceutics, 2017, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2017.07.039.

[154] Janos Szebeni, Gert Storm, Complement activation as a bioequivalence issue relevant to the development of generic liposomes and other nanoparticulate drugs, Biochemical and Biophysical Research Communications, Volume 468, Issue 3, 2015, Pages 490-497, ISSN 0006-291X, http://dx.doi.org/10.1016/j.bbrc.2015.06.177.

[155] Kaihua Chen, Jiancheng Guan, A bibliometric investigation of research performance in emerging nanobiopharmaceuticals, Journal of Informetrics, Volume 5, Issue 2, 2011, Pages 233-247, ISSN 1751-1577, http://dx.doi.org/10.1016/j.joi.2010.10.007.

[156] Raquel Requejo-Aguilar, Ana Alastrue-Agudo, Marta Cases-Villar, Eric Lopez-Mocholi, Richard England, María J. Vicent, Victoria Moreno-Manzano, Combined polymer-curcumin conjugate and ependymal progenitor/stem cell treatment enhances spinal cord injury functional recovery, Biomaterials, Volume 113, 2017, Pages 18-30, ISSN 0142-9612, http://dx.doi.org/10.1016/j.biomaterials.2016.10.032.

[157] Dmytro Golyshkin, Nazarii Kobyliak, Oleksandr Virchenko, Tetyana Falalyeyeva, Tetyana Beregova, Lyudmyla Ostapchenko, Martin Caprnda, Lubomir Skladany, Radka Opatrilova, Luis Rodrigo, Peter Kruzliak, Alexandr Shcherbokov, Mykola Spivak, Nanocrystalline cerium dioxide efficacy for prophylaxis of erosive and ulcerative lesions in the gastric mucosa of rats induced by stress, Biomedicine & Pharmacotherapy, Volume 84, 2016, Pages 1383-1392, ISSN 0753-3322, http://dx.doi.org/10.1016/j.biopha.2016.10.060.

[158] Aleksandra Szulc, Lukasz Pulaski, Dietmar Appelhans, Brigitte Voit, Barbara Klajnert-Maculewicz, Sugar-modified poly(propylene imine) dendrimers as drug delivery agents for cytarabine to overcome drug resistance, International Journal of Pharmaceutics, Volume 513, Issue 1, 2016, Pages 572-583, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2016.09.063.

[159] Paula S. Haddad, Tatiana M. Martins, Lília D'Souza-Li, Li M. Li, Konradin Metze, Randall L. Adam, Marcelo Knobel, Daniela Zanchet, Structural and morphological investigation of magnetic nanoparticles based on iron oxides for biomedical applications, Materials Science and Engineering: C, Volume 28, Issue 4, 2008, Pages 489-494, ISSN 0928-4931, http://dx.doi.org/10.1016/j.msec.2007.04.014.

[160] Serge Mignani, Sa?d El Kazzouli, Mosto Bousmina, Jean-Pierre Majoral, Dendrimer space concept for innovative nanomedicine: A futuristic vision for medicinal chemistry, Progress in Polymer Science, Volume 38, Issue 7, 2013, Pages 993-1008, ISSN 0079-6700, http://dx.doi.org/10.1016/j.progpolymsci.2013.03.003.

[161] Mike A.W. Eaton, Laurent Levy, Olivier M.A. Fontaine, Delivering nanomedicines to patients: A practical guide, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 11, Issue 4, 2015, Pages 983-992, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2015.02.004.

[162] Giovanna Lollo, Pablo Hervella, Pilar Calvo, Pablo Avilés, Maria Jose Guillén, Marcos Garcia-Fuentes, Maria José Alonso, Dolores Torres, Enhanced therapeutic efficacy of plitidepsin-loaded nanocapsules decorated with a new poly-aminoacid-PEG derivative, International Journal of Pharmaceutics, Volume 483, Issue 1, 2015, Pages 212-219, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2015.02.028.

[163] N. Thompson, M. Ahn, G. Chessari, K. Hearn, C.N. Johnson, J. Lewis, G. Ward, P. Williams, A. Woolford, 85 Characterization of a Potent XIAP and CIAP1 Dual Antagonist in Models of Melanoma and Leukemia, European Journal of Cancer, Volume 48, 2012, Page 27, ISSN 0959-8049, http://dx.doi.org/10.1016/S0959-8049(12)71883-X.

[164] Graphical Abstracts, Journal of Fluorine Chemistry, Volume 174, 2015, Pages vii-xii, ISSN 0022-1139, http://dx.doi.org/10.1016/S0022-1139(15)00114-1.

[165] Eameema Muntimadugu, Nagavendra Kommineni, Wahid Khan, Exploring the Potential of Nanotherapeutics in Targeting Tumor Microenvironment for Cancer Therapy, Pharmacological Research, 2017, ISSN 1043-6618, http://dx.doi.org/10.1016/j.phrs.2017.05.010.

[166] Marianna Foldvari, Mukasa Bagonluri, Carbon nanotubes as functional excipients for nanomedicines: II. Drug delivery and biocompatibility issues, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 4, Issue 3, 2008, Pages 183-200, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2008.04.003.

[167] Graphical Abstracts, Journal of Fluorine Chemistry, Volume 171, 2015, Pages vii-xiii, ISSN 0022-1139, http://dx.doi.org/10.1016/S0022-1139(15)00032-9.

[168] J.S. Riley, P.G. Johnston, D.B. Longley, 83 Investigation of Post-translational Modifications of c-FLIP, European Journal of Cancer, Volume 48, 2012, Page 27, ISSN 0959-8049, http://dx.doi.org/10.1016/S0959-8049(12)71881-6.

[169] Elisabete Fernandes, José Alexandre Ferreira, Peixoto Andreia, Lima Luís, Sérgio Barroso, Bruno Sarmento, Lúcio Lara Santos, New trends in guided nanotherapies for digestive cancers: A systematic review, Journal of Controlled Release, Volume 209, 2015, Pages 288-307, ISSN 0168-3659, http://dx.doi.org/10.1016/ j.jconrel.2015.05.003.

[170] Neelesh Kumar Mehra, Srinath Palakurthi, Interactions between carbon nanotubes and bioactives: a drug delivery perspective, Drug Discovery Today, Volume 21, Issue 4, 2016, Pages 585-597, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2015.11.011.

[171] Serge Mignani, Scot Huber, Helena Tomás, Jo?o Rodrigues, Jean-Pierre Majoral, Why and how have drug discovery strategies in pharma changed? What are the new mindsets?, Drug Discovery Today, Volume 21, Issue 2, 2016, Pages 239-249, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2015.09.007.

[172] Elenaz Naderkhani, Astrid Erber, Nata?a ?kalko-Basnet, G?ril Eide Flaten, Improved Permeability of Acyclovir: Optimization of Mucoadhesive Liposomes Using the Phospholipid Vesicle-Based Permeation Assay, Journal of Pharmaceutical Sciences, Volume 103, Issue 2, 2014, Pages 661-668, ISSN 0022-3549, http://dx.doi.org/10.1002/jps.23845.

[173] David Newton, Literature listing, World Patent Information, Volume 35, Issue 4, 2013, Pages 352-357, ISSN 0172-2190, http://dx.doi.org/10.1016/j.wpi.2013.06.006.

[174] Ichio Aoki, Misao Yoneyama, Jun Hirose, Yuzuru Minemoto, Takayoshi Koyama, Daisuke Kokuryo, Rumiana Bakalova, Shuhei Murayama, Tsuneo Saga, Sadahito Aoshima, Yukihito Ishizaka, Kenji Kono, Thermoactivatable polymer-grafted liposomes for low-invasive image-guided chemotherapy, Translational Research, Volume 166, Issue 6, 2015, Pages 660-673.e1, ISSN 1931-5244, http://dx.doi.org/10.1016/ j.trsl.2015.07.009.

[175] K.B. Ita, Transdermal drug delivery: progress and challenges, Journal of Drug Delivery Science and Technology, Volume 24, Issue 3, 2014, Pages 245-250, ISSN 1773-2247, http://dx.doi.org/10.1016/S1773-2247(14)50041-X.

[176] Je-Ruei Liu, Guo-Feng Chen, Hui-Nung Shih, Ping-Chung Kuo, Enhanced antioxidant bioactivity of (Danshen) products prepared using nanotechnology, Phytomedicine, Volume 15, Issue 1, 2008, Pages 23-30, ISSN 0944-7113, http://dx.doi.org/10.1016/j.phymed.2007.11.012.

[177] Surya K. Mallapragada, Timothy M. Brenza, JoEllyn M. McMillan, Balaji Narasimhan, Donald S. Sakaguchi, Anup D. Sharma, Svitlana Zbarska, Howard E. Gendelman, Enabling nanomaterial, nanofabrication and cellular technologies for nanoneuromedicines, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 11, Issue 3, 2015, Pages 715-729, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2014.12.013.

[178] Carina Peres, Ana I. Matos, Jo?o Conniot, Vanessa Sainz, Eva Zupan?i?, Joana M. Silva, Luís Gra?a, Rogério Sá Gaspar, Véronique Préat, Helena F. Florindo, Poly(lactic acid)-based particulate systems are promising tools for immune modulation, Acta Biomaterialia, Volume 48, 2017, Pages 41-57, ISSN 1742-7061, http://dx.doi.org/10.1016/j.actbio.2016.11.012.

[179] A.J. Ferreira, J. Cemlyn-Jones, C. Robalo Cordeiro, Nanoparticles, nanotechnology and pulmonary nanotoxicology, Revista Portuguesa de Pneumologia (English Edition), Volume 19, Issue 1, 2013, Pages 28-37, ISSN 2173-5115, http://dx.doi.org/10.1016/j.rppnen.2013.01.004.

[180] A.J. Ferreira, J. Cemlyn-Jones, C. Robalo Cordeiro, Nanoparticles, nanotechnology and pulmonary nanotoxicology, Revista Portuguesa de Pneumologia, Volume 19, Issue 1, 2013, Pages 28-37, ISSN 0873-2159, http://dx.doi.org/10.1016/j.rppneu.2012.09.003.

[181] Aurelio Salerno, Concepción Domingo Pascual, Bio-based polymers, supercritical fluids and tissue engineering, Process Biochemistry, Volume 50, Issue 5, 2015, Pages 826-838, ISSN 1359-5113, http://dx.doi.org/10.1016/ j.procbio.2015.02.009.

[182] Betty Tyler, David Gullotti, Antonella Mangraviti, Tadanobu Utsuki, Henry Brem, Polylactic acid (PLA) controlled delivery carriers for biomedical applications, Advanced Drug Delivery Reviews, Volume 107, 2016, Pages 163-175, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2016.06.018.

[183] Daniela Iannazzo, Alessandro Pistone, Signorino Galvagno, Stefania Ferro, Laura De Luca, Anna Maria Monforte, Tatiana Da Ros, Caroline Hadad, Maurizio Prato, Christophe Pannecouque, Synthesis and anti-HIV activity of carboxylated and drug-conjugated multi-walled carbon nanotubes, Carbon, Volume 82, 2015, Pages 548-561, ISSN 0008-6223, http://dx.doi.org/10.1016/j.carbon.2014.11.007.

[184] Anita Jemec, Petar Djinovi?, Tatjana Ti?ler, Albin Pintar, Effects of four CeO nanocrystalline catalysts on early-life stages of zebrafish and crustacean , Journal of Hazardous Materials, Volume 219, 2012, Pages 213-220, ISSN 0304-3894, http://dx.doi.org/10.1016/j.jhazmat.2012.03.080.

[185] H.L. Chen, W.T. Tai, C.W. Shiau, C.Y. Liu, C.S. Lin, A.L. Cheng, P.J. Chen, K.F. Chen, 82 Sorafenib and Its Derivative SC-59 Induces Autophagy in Hepatocellular Carcinoma Through SHP-1 Dependent Inhibition of STAT3, European Journal of Cancer, Volume 48, 2012, Pages 26-27, ISSN 0959-8049, http://dx.doi.org/10.1016/ S0959-8049(12)71880-4.

[186] N. Lütscher, S. H?nes, M. Grubert, M.E. Scheulen, R.A. Hilger, 86 Antitumoral Activity of a New Class of Triazenes, European Journal of Cancer, Volume 48, 2012, Pages 27-28, ISSN 0959-8049, http://dx.doi.org/ 10.1016/S0959-8049(12)71884-1.

[187] Chun-Woong Park, Xiaojian Li, Frederick G. Vogt, Don Hayes, Joseph B. Zwischenberger, Eun-Seok Park, Heidi M. Mansour, Advanced spray-dried design, physicochemical characterization, and aerosol dispersion performance of vancomycin and clarithromycin multifunctional controlled release particles for targeted respiratory delivery as dry powder inhalation aerosols, International Journal of Pharmaceutics, Volume 455, Issue 1, 2013, Pages 374-392, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2013.06.047.

[188] H. Huang, Q. Yuan, J.S. Shah, R.D.K. Misra, A new family of folate-decorated and carbon nanotube-mediated drug delivery system: Synthesis and drug delivery response, Advanced Drug Delivery Reviews, Volume 63, Issue 14, 2011, Pages 1332-1339, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2011.04.001.

[189] D. Depan, J. Shah, R.D.K. Misra, Controlled release of drug from folate-decorated and graphene mediated drug delivery system: Synthesis, loading efficiency, and drug release response, Materials Science and Engineering: C, Volume 31, Issue 7, 2011, Pages 1305-1312, ISSN 0928-4931, http://dx.doi.org/10.1016/j.msec.2011.04.010.

[190] Shutao Guo, Leaf Huang, Nanoparticles containing insoluble drug for cancer therapy, Biotechnology Advances, Volume 32, Issue 4, 2014, Pages 778-788, ISSN 0734-9750, http://dx.doi.org/10.1016/j.biotechadv.2013.10.002.

[191] Ruth Duncan, Polymer therapeutics as nanomedicines: new perspectives, Current Opinion in Biotechnology, Volume 22, Issue 4, 2011, Pages 492-501, ISSN 0958-1669, http://dx.doi.org/10.1016/j.copbio.2011.05.507.

[192] Nor Azwadi Che Sidik, Muhammad Noor Afiq Witri Muhammad Yazid, Syahrullail Samion, Mohamad Nor Musa, Rizalman Mamat, Latest development on computational approaches for nanofluid flow modeling: Navier–Stokes based multiphase models, International Communications in Heat and Mass Transfer, Volume 74, 2016, Pages 114-124, ISSN 0735-1933, http://dx.doi.org/10.1016/j.icheatmasstransfer.2016.03.007.

[193] Q. Yuan, S. Hein, R.D.K. Misra, New generation of chitosan-encapsulated ZnO quantum dots loaded with drug: Synthesis, characterization and in vitro drug delivery response, Acta Biomaterialia, Volume 6, Issue 7, 2010, Pages 2732-2739, ISSN 1742-7061, http://dx.doi.org/10.1016/j.actbio.2010.01.025.

[194] Weiwei He, Yitong Liu, Wayne G. Wamer, Jun-Jie Yin, Electron spin resonance spectroscopy for the study of nanomaterial-mediated generation of reactive Oxygen species, Journal of Food and Drug Analysis, Volume 22, Issue 1, 2014, Pages 49-63, ISSN 1021-9498, http://dx.doi.org/10.1016/j.jfda.2014.01.004.

[195] Jing An, Yuqiang Gou, Chunxia Yang, Fangdi Hu, Chunming Wang, Synthesis of a biocompatible gelatin functionalized graphene nanosheets and its application for drug delivery, Materials Science and Engineering: C, Volume 33, Issue 5, 2013, Pages 2827-2837, ISSN 0928-4931, http://dx.doi.org/10.1016/j.msec.2013.03.008.

[196] Samantha A. Meenach, Kimberly W. Anderson, J. Zach Hilt, Ronald C. McGarry, Heidi M. Mansour, Characterization and aerosol dispersion performance of advanced spray-dried chemotherapeutic PEGylated phospholipid particles for dry powder inhalation delivery in lung cancer, European Journal of Pharmaceutical Sciences, Volume 49, Issue 4, 2013, Pages 699-711, ISSN 0928-0987, http://dx.doi.org/10.1016/j.ejps.2013.05.012.

[197] Richard M. England, Esther Masiá, Vanessa Giménez, Rut Lucas, María J. Vicent, Polyacetal-stilbene conjugates — The first examples of polymer therapeutics for the inhibition of HIF-1 in the treatment of solid tumours, Journal of Controlled Release, Volume 164, Issue 3, 2012, Pages 314-322, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2012.08.017.

[198] Natassa Pippa, Maria Merkouraki, Stergios Pispas, Costas Demetzos, DPPC:MPOx chimeric advanced Drug Delivery nano Systems (chi-aDDnSs): Physicochemical and structural characterization, stability and drug release studies, International Journal of Pharmaceutics, Volume 450, Issue 1, 2013, Pages 1-10, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2013.03.052.

[199] Patrick Boisseau, Bertrand Loubaton, Nanomedicine, nanotechnology in medicine, Comptes Rendus Physique, Volume 12, Issue 7, 2011, Pages 620-636, ISSN 1631-0705, http://dx.doi.org/10.1016/j.crhy.2011.06.001.

[200] Oksana Petrichenko, Martins Rucins, Aleksandra Vezane, Irena Timofejeva, Arkadij Sobolev, Brigita Cekavicus, Karlis Pajuste, Mara Plotniece, Marina Gosteva, Tatjana Kozlovska, Aiva Plotniece, Studies of the physicochemical and structural properties of self-assembling cationic pyridine derivatives as gene delivery agents, Chemistry and Physics of Lipids, Volume 191, 2015, Pages 25-37, ISSN 0009-3084, http://dx.doi.org/10.1016/ j.chemphyslip.2015.08.005.

[201] Alicia Rodríguez-Gascón, Ana del Pozo-Rodríguez, Arantxazu Isla, María Angeles Solinís, Vaginal gene therapy, Advanced Drug Delivery Reviews, Volume 92, 2015, Pages 71-83, ISSN 0169-409X, http://dx.doi.org/10.1016/ j.addr.2015.07.002.

[202] Heico J. Frima, Cristina Gabellieri, Maj-Inger Nilsson, Drug delivery research in the European Union's Seventh Framework Programme for Research, Journal of Controlled Release, Volume 161, Issue 2, 2012, Pages 409-415, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2012.01.044.

[203] Murali M. Yallapu, Neeraj Chauhan, Shadi F. Othman, Vahid Khalilzad-Sharghi, Mara C. Ebeling, Sheema Khan, Meena Jaggi, Subhash C. Chauhan, Implications of protein corona on physico-chemical and biological properties of magnetic nanoparticles, Biomaterials, Volume 46, 2015, Pages 1-12, ISSN 0142-9612, http://dx.doi.org/10.1016/j.biomaterials.2014.12.045.

[204] Jinghua Duan, Heidi M. Mansour, Yangde Zhang, Xingming Deng, Yuxiang Chen, Jiwei Wang, Yifeng Pan, Jinfeng Zhao, Reversion of multidrug resistance by co-encapsulation of doxorubicin and curcumin in chitosan/poly(butyl cyanoacrylate) nanoparticles, International Journal of Pharmaceutics, Volume 426, Issue 1, 2012, Pages 193-201, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2012.01.020.

[205] Roman A. Perez, Hae-Won Kim, Core–shell designed scaffolds for drug delivery and tissue engineering, Acta Biomaterialia, Volume 21, 2015, Pages 2-19, ISSN 1742-7061, http://dx.doi.org/10.1016/j.actbio.2015.03.013.

[206] Luca Costantino, Diana Boraschi, Is there a clinical future for polymeric nanoparticles as brain-targeting drug delivery agents?, Drug Discovery Today, Volume 17, Issue 7, 2012, Pages 367-378, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2011.10.028.

[207] Chiming Wei, Yuri L. Lyubchenko, Hamid Ghandehari, Justin Hanes, Kathleen J. Stebe, Hai-Quan Mao, Donald T. Haynie, Donald A. Tomalia, Marianna Foldvari, Nancy Monteiro-Riviere, Petia Simeonova, Shuming Nie, Hidezo Mori, Susan P. Gilbert, David Needham, New technology and clinical applications of nanomedicine: Highlights of the second annual meeting of the American Academy of Nanomedicine (Part I), Nanomedicine: Nanotechnology, Biology and Medicine, Volume 2, Issue 4, 2006, Pages 253-263, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2006.11.001.

[208] James S. Murday, Richard W. Siegel, Judith Stein, J. Fraser Wright, Translational nanomedicine: status assessment and opportunities, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 5, Issue 3, 2009, Pages 251-273, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2009.06.001.

[209] Nishu Dixit, Kumar Vaibhav, Ravi Shankar Pandey, Upendra Kumar Jain, Om Prakash Katare, Anju Katyal, Jitender Madan, Improved cisplatin delivery in cervical cancer cells by utilizing folate-grafted non-aggregated gelatin nanoparticles, Biomedicine & Pharmacotherapy, Volume 69, 2015, Pages 1-10, ISSN 0753-3322, http://dx.doi.org/10.1016/j.biopha.2014.10.016.

[210] Hareesh B. Nair, Bokyung Sung, Vivek R. Yadav, Ramaswamy Kannappan, Madan M. Chaturvedi, Bharat B. Aggarwal, Delivery of antiinflammatory nutraceuticals by nanoparticles for the prevention and treatment of cancer, Biochemical Pharmacology, Volume 80, Issue 12, 2010, Pages 1833-1843, ISSN 0006-2952, http://dx.doi.org/10.1016/j.bcp.2010.07.021.

[211] Raj Bawa, S.R. Bawa, Stephen B. Maebius, Ted Flynn, Chiming Wei, Protecting new ideas and inventions in nanomedicine with patents, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 1, Issue 2, 2005, Pages 150-158, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2005.03.009.

[212] Samad Mussa Farkhani, Alireza Valizadeh, Hadi Karami, Samane Mohammadi, Nasrin Sohrabi, Fariba Badrzadeh, Cell penetrating peptides: Efficient vectors for delivery of nanoparticles, nanocarriers, therapeutic and diagnostic molecules, Peptides, Volume 57, 2014, Pages 78-94, ISSN 0196-9781. http://dx.doi.org/10.1016/ j.peptides.2014.04.015.

[213] Graphical Abstracts, Journal of Fluorine Chemistry, Volume 168, 2014, Pages v-xv, ISSN 0022-1139, http://dx.doi.org/10.1016/S0022-1139(14)00353-4.

[214] Ratnesh Lal, Morton F. Arnsdorf, Multidimensional atomic force microscopy for drug discovery: A versatile tool for defining targets, designing therapeutics and monitoring their efficacy, Life Sciences, Volume 86, Issue 15, 2010, Pages 545-562, ISSN 0024-3205, http://dx.doi.org/10.1016/j.lfs.2009.02.030.

[215] Fredrik Hacklin, Christian Marxt, Fritz Fahrni, Coevolutionary cycles of convergence: An extrapolation from the ICT industry, Technological Forecasting and Social Change, Volume 76, Issue 6, 2009, Pages 723-736, ISSN 0040-1625, http://dx.doi.org/10.1016/j.techfore.2009.03.003.

[216] Alberto A. Gabizon, Yogita Patil, Ninh M. La-Beck, New insights and evolving role of pegylated liposomal doxorubicin in cancer therapy, Drug Resistance Updates, Volume 29, 2016, Pages 90-106, ISSN 1368-7646, http://dx.doi.org/10.1016/j.drup.2016.10.003.

[217] Jianxiang Zhang, Peter X. Ma, Cyclodextrin-based supramolecular systems for drug delivery: Recent progress and future perspective, Advanced Drug Delivery Reviews, Volume 65, Issue 9, 2013, Pages 1215-1233, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2013.05.001.

[218] ?. Vani?, N. ?kalko-Basnet, Mucosal nanosystems for improved topical drug delivery: vaginal route of administration, Journal of Drug Delivery Science and Technology, Volume 24, Issue 5, 2014, Pages 435-444, ISSN 1773-2247, http://dx.doi.org/10.1016/S1773-2247(14)50085-8.

[219] Rutledge Ellis-Behnke, Nano Neurology and the Four P's of Central Nervous System Regeneration: Preserve, Permit, Promote, Plasticity, Medical Clinics of North America, Volume 91, Issue 5, 2007, Pages 937-962, ISSN 0025-7125, http://dx.doi.org/10.1016/j.mcna.2007.04.005.

[220] Kale Mohana Raghava Srivalli, Brahmeshwar Mishra, Drug nanocrystals: A way toward scale-up, Saudi Pharmaceutical Journal, Volume 24, Issue 4, 2016, Pages 386-404, ISSN 1319-0164, http://dx.doi.org/10.1016/ j.jsps.2014.04.007.

[221] Eva-Maria Collnot, Hussain Ali, Claus-Michael Lehr, Nano- and microparticulate drug carriers for targeting of the inflamed intestinal mucosa, Journal of Controlled Release, Volume 161, Issue 2, 2012, Pages 235-246, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2012.01.028.

[222] Joshua J. Rychak, Jonathan R. Lindner, Klaus Ley, Alexander L. Klibanov, Deformable gas-filled microbubbles targeted to P-selectin, Journal of Controlled Release, Volume 114, Issue 3, 2006, Pages 288-299, ISSN 0168-3659, http://dx.doi.org/10.1016/j.jconrel.2006.06.008.

[223] Cezary Watala, Kamil Karolczak, Hassan Kassassir, Marcin Talar, Tomasz Przygodzki, Katarzyna Maczynska, Magdalena Labieniec-Watala, How do the full-generation poly(amido)amine (PAMAM) dendrimers activate blood platelets? Activation of circulating platelets and formation of “fibrinogen aggregates” in the presence of polycations, International Journal of Pharmaceutics, Volume 503, Issue 1, 2016, Pages 247-261, ISSN 0378-5173, http://dx.doi.org/10.1016/j.ijpharm.2015.08.073.

[224] M.S. Palombo, Y. Singh, P.J. Sinko, Prodrug and conjugate drug delivery strategies for improving HIV/AIDS therapy, Journal of Drug Delivery Science and Technology, Volume 19, Issue 1, 2009, Pages 3-14, ISSN 1773-2247, http://dx.doi.org/10.1016/S1773-2247(09)50001-9.

[225] Table of Contents, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 7, Issue 4, 2011, Pages A4-A5, ISSN 1549-9634, http://dx.doi.org/10.1016/S1549-9634(11)00192-4.

[226] Andriy Kuzmov, Tamara Minko, Nanotechnology approaches for inhalation treatment of lung diseases, Journal of Controlled Release, Volume 219, 2015, Pages 500-518, ISSN 0168-3659, http://dx.doi.org/10.1016/ j.jconrel.2015.07.024.

[227] Yolanda Diebold, Margarita Calonge, Applications of nanoparticles in ophthalmology, Progress in Retinal and Eye Research, Volume 29, Issue 6, 2010, Pages 596-609, ISSN 1350-9462, http://dx.doi.org/10.1016/ j.preteyeres.2010.08.002.

[228] Ramazan Bal, Gaffari Türk, Mehmet Tuzcu, Okkes Yilmaz, Ibrahim Ozercan, Tuncay Kuloglu, Seyfettin Gür, Victor S. Nedzvetsky, Artem A. Tykhomyrov, Grigory V. Andrievsky, Giyasettin Baydas, Mustafa Naziroglu, Protective effects of nanostructures of hydrated C fullerene on reproductive function in streptozotocin-diabetic male rats, Toxicology, Volume 282, Issue 3, 2011, Pages 69-81, ISSN 0300-483X, http://dx.doi.org/10.1016/ j.tox.2010.12.003.

[229] Dhruba J. Bharali, Shaker A. Mousa, Emerging nanomedicines for early cancer detection and improved treatment: Current perspective and future promise, Pharmacology & Therapeutics, Volume 128, Issue 2, 2010, Pages 324-335, ISSN 0163-7258, http://dx.doi.org/10.1016/j.pharmthera.2010.07.007.

[230] Sandipan Ray, Harini Chandra, Sanjeeva Srivastava, Nanotechniques in proteomics: Current status, promises and challenges, Biosensors and Bioelectronics, Volume 25, Issue 11, 2010, Pages 2389-2401, ISSN 0956-5663, http://dx.doi.org/10.1016/j.bios.2010.04.010.

[231] B. Mishra, Bhavesh B. Patel, Sanjay Tiwari, Colloidal nanocarriers: a review on formulation technology, types and applications toward targeted drug delivery, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 6, Issue 1, 2010, Pages 9-24, ISSN 1549-9634, http://dx.doi.org/10.1016/j.nano.2009.04.008.

[232] Contents, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 4, Issue 4, 2008, Pages A3-A4, ISSN 1549-9634, http://dx.doi.org/10.1016/S1549-9634(08)00178-0.

[233] Vladimir Torchilin, Multifunctional and stimuli-sensitive pharmaceutical nanocarriers, European Journal of Pharmaceutics and Biopharmaceutics, Volume 71, Issue 3, 2009, Pages 431-444, ISSN 0939-6411, http://dx.doi.org/10.1016/j.ejpb.2008.09.026.

[234] Francesca A. Cupaioli, Fabio A. Zucca, Diana Boraschi, Luigi Zecca, Engineered nanoparticles. How brain friendly is this new guest?, Progress in Neurobiology, Volume 119, 2014, Pages 20-38, ISSN 0301-0082, http://dx.doi.org/10.1016/j.pneurobio.2014.05.002.

[235] Alejandro Sosnik, Angel M. Carcaboso, Nanomedicines in the future of pediatric therapy, Advanced Drug Delivery Reviews, Volume 73, 2014, Pages 140-161, ISSN 0169-409X, http://dx.doi.org/10.1016/ j.addr.2014.05.004.

[236] Table of Contents, Nanomedicine: Nanotechnology, Biology and Medicine, Volume 10, Issue 1, 2014, Pages A4-A7, ISSN 1549-9634, http://dx.doi.org/10.1016/S1549-9634(13)00599-6.

[237] Jiancheng Guan, Qingjun Zhao, The impact of university–industry collaboration networks on innovation in nanobiopharmaceuticals, Technological Forecasting and Social Change, Volume 80, Issue 7, 2013, Pages 1271-1286, ISSN 0040-1625, http://dx.doi.org/10.1016/j.techfore.2012.11.013.

[238] Lisa C. Du Toit, Thirumala Govender, Trevor Carmichael, Pradeep Kumar, Yahya E. Choonara, Viness Pillay, Design of an Anti-Inflammatory Composite Nanosystem and Evaluation of Its Potential for Ocular Drug Delivery, Journal of Pharmaceutical Sciences, Volume 102, Issue 8, 2013, Pages 2780-2805, ISSN 0022-3549, http://dx.doi.org/10.1002/jps.23650.

[239] Emerging Fields, Free Radical Biology and Medicine, Volume 43, 2007, Pages S67-S74, ISSN 0891-5849, http://dx.doi.org/10.1016/j.freeradbiomed.2007.10.018.

[240] Xue-Qing Zhang, Xiaoyang Xu, Nicolas Bertrand, Eric Pridgen, Archana Swami, Omid C. Farokhzad, Interactions of nanomaterials and biological systems: Implications to personalized nanomedicine, Advanced Drug Delivery Reviews, Volume 64, Issue 13, 2012, Pages 1363-1384, ISSN 0169-409X, http://dx.doi.org/10.1016/ j.addr.2012.08.005.

[241] Arunachalam Muthaiyan, Alya Limayem, Steven C. Ricke, Antimicrobial strategies for limiting bacterial contaminants in fuel bioethanol fermentations, Progress in Energy and Combustion Science, Volume 37, Issue 3, 2011, Pages 351-370, ISSN 0360-1285, http://dx.doi.org/10.1016/j.pecs.2010.06.005.

[242] Scientific Programme – Details, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Pages xxiv-lxvi, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)71697-9.

[243] Subject Index, European Journal of Cancer Supplements, Volume 8, Issue 7, 2010, Pages 233-243, ISSN 1359-6349, http://dx.doi.org/10.1016/S1359-6349(10)72373-9.

[244] Ruth Duncan, Development of HPMA copolymer–anticancer conjugates: Clinical experience and lessons learnt, Advanced Drug Delivery Reviews, Volume 61, Issue 13, 2009, Pages 1131-1148, ISSN 0169-409X, http://dx.doi.org/10.1016/j.addr.2009.05.007.

[245] R. Palao-Suay, L.G. Gómez-Mascaraque, M.R. Aguilar, B. Vázquez-Lasa, J. San Román, Self-assembling polymer systems for advanced treatment of cancer and inflammation, Progress in Polymer Science, Volume 53, 2016, Pages 207-248, ISSN 0079-6700, http://dx.doi.org/10.1016/j.progpolymsci.2015.07.005.

[246] K. John Morrow, Raj Bawa, Chiming Wei, Recent Advances in Basic and Clinical Nanomedicine, Medical Clinics of North America, Volume 91, Issue 5, 2007, Pages 805-843, ISSN 0025-7125, http://dx.doi.org/10.1016/ j.mcna.2007.05.009.

[247] Tissue/Cell Targets and Reactions, Free Radical Biology and Medicine, Volume 41, 2006, Pages S144-S154, ISSN 0891-5849, http://dx.doi.org/10.1016/j.freeradbiomed.2006.10.015.

[248] Inflammation, Free Radical Biology and Medicine, Volume 41, 2006, Pages S68-S78, ISSN 0891-5849, http://dx.doi.org/10.1016/j.freeradbiomed.2006.10.008.

[249] Vinay Deep Punetha, Sravendra Rana, Hye Jin Yoo, Alok Chaurasia, James T. McLeskey, Madeshwaran Sekkarapatti Ramasamy, Nanda Gopal Sahoo, Jae Whan Cho, Functionalization of carbon nanomaterials for advanced polymer nanocomposites: A comparison study between CNT and graphene, Progress in Polymer Science, Volume 67, 2017, Pages 1-47, ISSN 0079-6700, http://dx.doi.org/10.1016/j.progpolymsci.2016.12.010.

[250] Antioxidants, Nutrition & Health, Free Radical Biology and Medicine, Volume 41, 2006, Pages S18-S31, ISSN 0891-5849, http://dx.doi.org/10.1016/j.freeradbiomed.2006.10.002.

[251] Scientific Programme – Proffered Papers, European Journal of Cancer, Volume 49, 2013, Pages S97-S153, ISSN 0959-8049, http://dx.doi.org/10.1016/S0959-8049(13)70060-1.

[252] Subject Index, European Journal of Cancer, Volume 49, 2013, Pages S975-S1028, ISSN 0959-8049, http://dx.doi.org/10.1016/S0959-8049(13)70067-4.

[253] Goldschmidt Abstracts 2010 – H, Geochimica et Cosmochimica Acta, Volume 74, Issue 12, 2010, Pages A369-A440, ISSN 0016-7037, http://dx.doi.org/10.1016/j.gca.2010.04.033.

[254] Sivakumar Manickam, Editorial Note, Ultrasonics Sonochemistry, Volume 35, 2017, Pages 529-530, ISSN 1350-4177, http://dx.doi.org/10.1016/j.ultsonch.2016.06.028.

[255] Juan G. Osorio, Fernando J. Muzzio, Evaluation of resonant acoustic mixing performance, Powder Technology, Volume 278, 2015, Pages 46-56, ISSN 0032-5910, http://dx.doi.org/10.1016/j.powtec.2015.02.033.

[256] Zahra Karami, Mehrdad Hamidi, Cubosomes: remarkable drug delivery potential, Drug Discovery Today, Volume 21, Issue 5, 2016, Pages 789-801, ISSN 1359-6446, http://dx.doi.org/10.1016/j.drudis.2016.01.004.

[257] Jungil Park, Hyunwook Nam, Sun Young Ahn, Youngmi Kim Pak, James Jungho Pak, A reservoir-type oxygen sensor with 2×3 array for measuring cellular respiration levels, Sensors and Actuators B: Chemical, Volume 176, 2013, Pages 913-920, ISSN 0925-4005, http://dx.doi.org/10.1016/j.snb.2012.09.037.