Accepted Manuscript Title: An Insight into functionalized Calcium based Inorganic Nanomaterials in Biomedicine: Trends and Transitions Author: Shweta Sharma Ashwni verma B. Venkatesh Teja Gitu Pandey Naresh Mittapelly Ritu Trivedi P.R. Mishra PII: DOI: Reference:

S0927-7765(15)00311-2 http://dx.doi.org/doi:10.1016/j.colsurfb.2015.05.014 COLSUB 7087

To appear in:

Colloids and Surfaces B: Biointerfaces

Received date: Revised date: Accepted date:

14-11-2014 6-5-2015 8-5-2015

Please cite this article as: S. Sharma, A. verma, B.V. Teja, G. Pandey, N. Mittapelly, R. Trivedi, P.R. Mishra, An Insight into functionalized Calcium based Inorganic Nanomaterials in Biomedicine: Trends and Transitions, Colloids and Surfaces B: Biointerfaces (2015), http://dx.doi.org/10.1016/j.colsurfb.2015.05.014 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Highlights of the present review

2

5

 Focus is more on systemic applications than localized delivery

6

 Discussion on application in gene, protein and small molecules delivery for diseases like cancer, osteoporosis, diabetes and vaccination.

9

cr

8

 Covers all the surface modification or functionalization that has been done to improve their applicability.

us

7

ip t

4

 Discussion on characteristic properties in comparison to other nanomaterials and method of preparation

3

12

 Discussion on Major challenges in clinical application.

M

an

11

 Mineralization of calcium phosphate over other nanosized delivery systems

10

13

17

te

16

Ac ce p

15

d

14

1 Page 1 of 47

17 19 20 21 22 23 24

An Insight into functionalized Calcium based Inorganic Nanomaterials in Biomedicine: Trends and Transitions Shweta Sharma1, Ashwni verma1, B Venkatesh Teja1, Gitu Pandey1, Naresh Mittapelly1, Ritu Trivedi2, P.R.Mishra1* 1- Division of Pharmaceutics, CSIR-Central Drug Research Institute

ip t

18

2-Division of Endocrinology, CSIR-Central Drug Research Institute

26

B 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow (U.P.) 226031

27 28 29 30 31 32 33 34 35 36 37 38 39 40 41

*Corresponding Author Dr. P.R. Mishra Ph.D Division of Pharmaceutics, Preclinical South PCS 002/011, CSIR-Central Drug Research Institute B.S. 10/1, Sector-10, Jankipuram Extension, Sitapur Road, Lucknow-226031, India Phone 91-522-2772450 (4537) Fax: 91-522-2771941 E-mail: [email protected] [email protected]

us

an

M

d

te

Ac ce p

42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58

cr

25

2 Page 2 of 47

Abstract

Ac ce p

te

d

M

an

us

cr

ip t

Over the recent years the use of biocompatible and biodegradable nanoparticles in biomedicine has become a significant priority. Calcium based ceramic nanoparticles like calcium phosphate (CaP) and calcium carbonate (CaCO3) are therefore considered as attractive carriers as they are naturally present in human body with nanosize range. Their application in tissue engineering and localized controlled delivery of bioactives for bones and teeth is well established now, but recently their use has increased significantly as carrier of bioactives through other routes also. These delivery systems have become most potential alternatives to other commonly used delivery system because of their cost effectiveness, biodegradability, chemical stability, controlled and stimuli responsive behaviour. This review comprehensively covers their characteristic features, method of preparation and applications but the thrust is to focus their recent development, functionalization and use in systemic delivery. On the same platform mineralization of other nanoparticulate delivery system which has widened their application drug delivery will be discussed. The emphasis has been given on their pH dependent properties which make them excellent carriers for tumor targeting and intracellular delivery. Finally this review also attempts to discuss their drawback which limits their clinical utility.

Page 3 of 47

59

te

d

M

an

us

cr

ip t

1. Introduction: The rapid growth of nanotechnology in the last 35 years of therapeutics can be seen from the fact that the several products of nano-formulations like Caelyx®, Doxil®, Transdrug® , Abraxane® etc are already there in the market and apart from that there is an extensive list of systems which are in clinical trials or being scientifically explored. The reason behind this unprecedented growth of applications in the area of nanoscience and nanotechnology is the important and unique capabilities of nanosystems that are not found in the bulk sample of the same system. It mainly includes their large surface area, intracellular delivery and quantum properties. Nanomaterials have been used in almost every field of biomedical research, such as targeted delivery vehicles for therapeutic agents, contrast agents, biosensors and artificial oxygen carriers. The use of nanomaterial in the delivery of therapeutics provides liberty to modify their fundamental properties such as diffusivity, solubility, drug release characteristics, blood circulation half-life, bio-distribution, metabolism and immunogenicity. Further, tailoring their surface properties allows temporal and site specific delivery of therapeutics useful in the treatment of variety of diseases and disorders [1]. Nanosized delivery systems used in biomedical research can widely be classified as polymeric nanoparticles, polysaccharide nanoparticles, protein nanoparticles, dendrimers, nanoshells, micelles, engineered viral nanoparticles and metallic nanoparticles. These systems have shown potential for several branches of medicine such as immunology, neurology, oncology, cardiology, endocrinology, pulmonary, ophthalmology, orthopedics and dentistry. Polymeric and lipidic nanoparticles are called as “soft” nanoparticles and currently the most employed systems because of their biocompatible and biodegradable nature. On the other side inorganic particles like metallic and ceramic nanoparticles are known as “hard” nanoparticles because of their dense nature. In the current scenario of biomedicine application of inorganic nanoparticles has advanced rapidly and extensive amount of work is being done in their synthesis and surface modification. Inorganic nanoparticles covers a wide range of materials and therefore can further be classified as metallic nanoparticles or ceramic nanoparticles. Metallic nanoparticles are the ones which are made up of metals like gold , silver or iron oxide whereas ceramic nanoparticles are made up of alumina (Al2O3), calcium phosphate (CaP), silica (SiO2), zirconia (ZrO2), titanium oxide (TiO2) or calcium carbonate (CaCO3) [2]. In the list of inorganic nanoparticles, calcium based ceramic nanoparticles are particularly gaining the interest of researchers as they can be synthesized easily with desired size and porosity and possess properties that other systems lack. Though, they have been in use for localized delivery from several decades, however; their systemic and targeted delivery is a recent topic of interest among researchers. The aim of the present review is to elaborately discuss their properties, method of preparation, applications and related key issues which need to be resolved. The review has been covered with most of the reports that have been published till date.

Ac ce p

60 61 62 63 64 65 66 67 68 69 70 71 72 73 74 75 76 77 78 79 80 81 82 83 84 85 86 87 88 89 90 91 92 93 94 95 96 97 98 99 100

2. Constraints of other frequently used nano-particulate delivery systems

3 Page 4 of 47

For the past several decades polymers, lipids and proteins are the most commonly used source materials for the development of nanoparticulate delivery systems. Even being potential drug carriers they have their own sort of troubles and limitations. Most challenging limitation with the commonly employed polymeric and lipidic systems is the usage of organic solvents during their synthesis leading to a big concern for their in-vivo applications. Moreover, delivery systems like Liposomes, micelles and emulsions suffer the limitation of stability as well as leakage of drug molecules in systemic circulation before reaching to target side. Though the limitations have been overcome to a certain extent by the use of polymeric nanoparticles like PLGA (FDA approved polymer) but it is also known to produce acidic products on their degradation which may have unwanted side effects on long term administration. Further, most of these delivery systems are not cost effective and lack industrial scalability. Cationic delivery systems commonly used in siRNA or pDNA delivery are toxic in nature and promote unwanted uptake by reticuloendothelial systems (RES). This review discusses how these limitations can be overcome to a certain extent by the use of CaP and CaCO3 nanoparticles.

138 139 140 141 142 143

Ceramic nanoparticles made up of CaP and CaCO3 are completely biocompatible and biodegradable compared to any other nanoparticles. Unlike carbon nanotubes, quantum dots, magnetic nanoparticles, silica nanoparticles and metallic nanoparticles they are free of severe toxicity. Metallic nanoparticles like Cr, Cd, Au, Ag, Se, Te,Co,TiO2, CuO and ZnO have been shown to increase mutation frequency, produce oxidative lesions, decrease cell viability and induce damage to DNA. Contrary to this calcium based nanosized carriers belong to the

an

us

cr

ip t

101 102 103 104 105 106 107 108 109 110 111 112 113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129 130 131 132 133 134 135 136 137

Ac ce p

te

d

M

3. “Nano-structured” CaP and CaCO3 particles: Idea behind development The keen observation of researchers that had attracted great interest of formulation scientists in developing mineralized biologically inspired delivery systems for therapeutics is that in biological systems most of the biomaterials such as bones, teeth or shells are composed of regular hierarchical structure of calcium phosphate and calcium carbonate. These structures have much superior mechanical and functional properties compared to the artificial materials. So it was believed that development of delivery vehicles composed of natural bio-mineral would possess an excellent biocompatibility owing to their chemical similarity to human hard tissues (bones, teeth) and ions of intracellular signalling pathways. Moreover, they are already present in blood at relative high concentration. It was also observed that biologically formed CaP and CaCO3 were often nanocrystals (5-20 nm width by 60 nm length) that are precipitated under mild conditions and this natural selection as nanostructured materials provides them the capability of specific interactions with natural proteins. Hence the development of nanosized bioresorbable inorganic materials such as CaP and CaCO3 in biomedical research stands to benefit most. In fact, their use in hard tissue engineering and as a matrix for localized controlled delivery of therapeutic agents to bones and tissues is very well established [3]. But nowadays they are also being explored for systemic delivery of therapeutics and this is because of their several attractive features (listed below) over others which makes them excellent delivery systems. 3.1. Biocompatibility and biodegradability

4 Page 5 of 47

ip t

safest class of materials known so far where by-products like Ca2+, PO43-or CO32-are already present in the bloodstream within the concentration range of 1-5mM [4]. If fabricated appropriately they can possess the same chemistry, size and crystalline structure as the targeted tissues which enhance the material’s activity and bio-acceptability before releasing the drug. Calcium phosphate per se belongs to a category of GRAS (Generally Regarded as Safe) as reported by FDA [5]

M

an

us

cr

3.2. In-vivo Stability In-vivo stability refers to the stability of nanoparticles in biological matrix. In this respect most of the inorganic nanoparticles bears the superiority over their counterparts and thus are relatively much more suitable for in-vivo applications. For example, some of the polymeric systems swell or their porosity changes on change of the pH and temperature while these nanoparticles do not. Also, they overcome the major limitation of micelle and liposomes based carriers, which are subjected to dissipation at below specific critical concentration (a major obstacle in in-vivo administration). CaP and CaCO3 nanoparticles are much more robust, particularly those with Ca/P ratio equal to hydroxyapatite (HAP, naturally occurring calcium phosphate) which is not soluble in blood as blood is itself supersaturated with respect to HAP. Also polymers like PLGA and PLLA inhibit rapid or prompt release of the loaded drugs and undergo catalytic degradation with time leading to the generation of acidic products. These ceramic nanoparticles are not prone to microbial growth, thus its storage stability is good.

te

d

3.3. Ease of preparation and cost effectiveness The simplicity in the manufacturing process with minimal raw materials (common salts) without the use of organic solvents makes them more cost effective and industrially relevant compared to other nanoparticulate systems. Convenience in fabrication of these nanoparticles in aqueous media provides a suitable platform for the surface functionalization with appropriate agents like siRNA, proteins and several polyelectrolytes.

Ac ce p

144 145 146 147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186

3.4. Particles in very small size range Their synthesis under controlled conditions can result in particles with size range HepG2 > HeLa but had no toxicity on normal hepatic cell line (L-02)[118]. Just like CaP nanoparticles CaCO3 nanoparticles are also potential candidates for drug delivery particularly due to their porous nature. Several modifications have been done to improve their usage and applicability for in vitro and in vivo purpose. Carboxy Methyl Chitosan (CMC) modified CaCO3 micro and nanoparticles showed 60 % encapsulation efficiency of doxorubicin (DOX) because of its porous structure and electrostatic interaction between negatively charged CMC and positively charged DOX [62]. Low molecular weight drug betamethasone or bioactive protein loaded CaCO3 nanoparticles showed enhanced chemical stability and sustained release on subcutaneous injection [122]. The nanoparticles also show potential application as calcium supplement where enhanced serum calcium concentrations has been achieved than conventional formulations [123]. 5.4.2. Ocular hypotensive agents delivery Apart from the delivery of anticancer drugs the other major area where CaP nanoparticles have been explored are in delivery of ocular hypotensive agents like timolol [119], 7hydroxy-2-dipropyl-aminotetralin [120] and methazolamide [121] with the major aim is to sustain the drug levels in anterior tissue for a period of few hours and reduce the dosing frequency as compared to the solution. Unlike CaP nanoparticles CaCO3 nanoparticles have not been investigated much for systemic delivery.

Ac ce p

737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780

5.5. In-vitro/ in-vivo imaging and photodynamic therapy Diagnosis of particular disease is very essential and for that several imaging methods have been in practice. The broad spectrum profile, photochemical instability and low bleaching threshold restricts the direct use of organic dyes and lanthanide chelates for bio-sensing and imaging purposes[124]. Quantum dots which are stable and produce bright fluorescence are frequently accepted as an alternative but their heavy metal toxicity limits their in-vivo use. Nanocarriers which can encapsulate fluorescent molecules in their rigid matrix of colloidal carrier can be a better substitute to shields interaction of dyes with solvent molecules as well as to improve photostability and in vivo stability [125]. The use of surface decorated particulate carriers for diagnostic imaging gives the additional advantage of cellular targeting and therapeutic activity. The encapsulation of fluorophores in CaP/CaCO3 nanoparticulate systems retains the stability associated with silica and polymeric nanoparticles but simultaneously eliminates the problems associated with other systems and therefore results in bright and stable fluorescent particles like quantum dots. Several reports are available CaP nanoparticles have been used as carriers for fluorescent probes either by doping with lanthanides or by surface functionalizing the organic dyes. CaP nanoparticles doped with lanthanide were prepared by precipitation method at low temperature and stabilised with 20 Page 21 of 47

te

d

M

an

us

cr

ip t

DNA to give a stable colloid. These particles were tested on in vitro HUVEC cell where much enhanced uptake of particles were observed by confocal laser scanning microscopy. In this study though the results were good but some morphological changes also observed which raises the concern regarding future use of lanthanide doped particles on in-vivo use due to potential toxicity[126, 127]. In another study conducted by Erhan I˙. Altınogˇ lu et al Indocyanine green (ICG, NIR emitting fluorophore) doped PEGylated CaP nanoparticles prepared for in vivo studies of breast adenocarcinoma tumors. The dye loaded particles showed 2-fold higher quantum efficiency per molecule and photostability which was 4.7-fold longer relative to free molecule making these ICG doped particles an attractive fluoroprobe for sensitive diagnostic imaging applications[128]. Number of other dyes such as rhodamine WT, fluorscein, Cy3 loaded particles have also been developed along with drugs for simultaneous imaging and drug delivery. Just like encapsulation surface functionalization is also an approach for loading of actives in CaP nanoparticles than involves either incorporation in coating layers or stabilisation by the actives only. In 2008 Kathirvel Ganesan et al prepared CaP nanoparticles surface functionalized with water soluble p-TPPP (5,10,15,20-tetrakis(4-phosphonooxyphenyl) porphine dye for photodynamic therapy in cancer. Photodynamic therapy is a technique useful in the treatment of tumours and bacterial biofilms in which light-sensitive dye is brought into the malignant tissue and irradiated with a laser source. The excited dye destroys the malignant cells or bacteria by forming singlet oxygen species. The goal of the study was to demonstrate that surface functionalization of organic dyes on CaP can also be a biocompatible and harmless technique for the delivery of fluorescing molecules[129]. They showed enhanced uptake of particles into NIH 3T3 fibroblast cells after a few hours of administration. Organic dyes can also be loaded on to polymeric shell decorated nanoparticles. Epple and co-workers did the experimental analysis by loading m-THPP (porphyrin dye) dye and methylene blue dye into the polymeric shell functionalized CaP nanoparticles. The final charge of the particle was adjusted by selecting an appropriate polymer. The efficiency of these positively charged nanoparticles were compared to free dye on HT- 22(human colon adenocarcinoma cells), HIG-82 (synoviocytes from rabbits) and J774A.1 cells (murine macrophages). The different activity profile was obtained depending upon the cell line, like for J774 A the particles were toxic even in dark and moderate activity was observed for HT29 epithelial cells but contrastingly particles showed a good performance with HIG-82 synoviocytes[130]. Thus though they were able to prepare water-dispersible system for dye by avoiding the alcoholic solution, the final efficiency/activity was dependent upon the number of other parameters, e.g. particle charge, kind of polymer, types of cell and cell culture medium (e.g. the presence of proteins) which needs further studies for complete justification[131]. Incorporation of a pH sensitive dye SNARF-1 and MRI probes in CaP nanoparticles has further widened the application of CaP nanoparticles in imaging[132]. Peng Mi et al observed higher amount of contrast agent at tumor position after intravenous injection of diethylenetriaminepentaacetic acid gadolinium(III) Gd-DTPA (an MRI probe) encapsulated CaP nanoparticles than free GdDTPA which was due to EPR effect leading to higher tumor accumulation as well as higher molecular relaxivity of Gd-DTPA than the free Gd-DTPA [133]. Recently, Yu-Cheng Tseng et al reported how lymphotropism can be achieved by LCP with particle size ~25nm. Intravenous administration of Indium (111In) loaded LCP by SPECT/CT imaging showed

Ac ce p

781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801 802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824

21 Page 22 of 47

ip t

~70% ID/g accumulation in lymph nodes compared to ~25% ID/g accumulation in liver and spleen. However they also found that particles with size >67nm were less lymphotropic. These particles were sufficiently able to visualize 4T1 breast cancer lymph node metastasis model [134]. Potential of CaP nanoparticles have also been shown through multi-modal imaging where combined delivery of nuclear (99m-Technetium-methylene diphosphonate 99mTc-MDP), magnetic (Gadolinium Gd3+) and near infrared imaging (indocyanine green ICG) in in-vivo models without showing any toxicity on healthy human mononuclear cells, red-blood cells and platelets [135].

te

d

M

an

us

cr

5.6. Multifunctional nanoparticles A recent advancement in nanomedicine-mediated therapy is the development of multifunctional carriers which involves combined delivery of two or more active molecules to serve the dual purpose. In fact these “multifunctional nanoparticles” have become a hot topic in recent times for biomedical applications. So far several promising systems for codelivery have been developed based on liposomes, polymeric/lipidic and silica-based nanoparticles[83]. Advantage of co-delivery of a gene and drug is that it can overcome the multidrug resistance (MDR) to main line drug and thus can increase its therapeutic efficacy. Just like other nanosized CaP and CaCO3 has also been investigated for simultaneous delivery of multiple molecules and as expected in this arena also they had proven themselves as potential candidates. On in vivo administration Alginate and KALA peptide modified CaCO3 nanoparticles showed enhanced tumor cell apoptosis on simultaneous incorporation of Doxorubicin (DOX) and gene plasmid (pGL3–Luc) compared to unmodified nanoparticles. The observed enhanced efficacy with Alginate modified CaCO3 nanoparticles was because of decreased size and improved physical stability [136] whereas better activity observed with KALA modified particle was because of endosomolytic and fusogenic property of the peptide [137, 138]. Another example of such includes the combined incorporation of cytotoxic drug (PTX, DOX) and fluorophores like Rhodamine-WT (Rh-WT)/fluorescein for theranostic purpose. A research group in The Pennsylvania State University used microemulsion approach to encapsulate the Cer6 and fluorescein in CaP nanoparticles. The nanoparticles induced 80% growth inhibition in human vascular smooth muscle cells at 25 fold lower concentration than ceramide solution in DMSO. The same research group also prepared nanoparticles loaded with Cer10 and WT rhodamine and performed in vitro studies in UACC 903 melanoma cells. Results revealed 5% lower survival of melanoma cells as compared to free drug dissolved in DMSO at a concentration of 5µM. Also no morphological changes were observed in control wells (particle without dye and ceramide) in both cases implying that blank particle themselves were not toxic. The Cer10 doped particles were also found to be significantly effective in sensitive as well as resistant MCF-7 breast cancer cell lines [139, 140]. Feng Chen et al developed dual doped Eu3+ (NIR probe) and Gd3+ (MRI probe) CaP vesicle like nanospheres in the presence of PLA-mPEG amphiphillic block copolymer as multifunctional delivery system for in-vivo bio-imaging and therapeutic activity. The particles were found non-toxic in in-vitro. Further the particle when loaded with drug (ibuprofen as a model drug) they observed that release of drug was sustained for a very long time with more than 80 days [141].Gemcitabine monophosphate and c-myc siRNA codelivery by anisamide conjugated LCP nanoparticles triggered apoptosis in ~28% of H460

Ac ce p

825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868

22 Page 23 of 47

subcutaneous and A549 orthotopic xenografts whereas only drug and only siRNA loaded LCP triggered apoptosis in ~ 23 % and ~ 3 % tumor cells respectively. Tumour volume was also found to be significantly less compared to the control on treatment with co-loaded nanoparticles [142].

an

us

cr

ip t

5.7. As inert templates for polyelectrolyte capsule The calcium based inorganic microparticles or nanoparticles can also be used as inert templates for the preparation of drug loaded hollow nanocapsules in place of particles like polystyrene latex and gold which are not biocompatible and biodegradable [106] [105]. Basically in this technique CaP or CaCO3 nanoparticles can serve as core template which carries the bioactive molecule and over which alternate layers of oppositely charged polymers are deposited as per the necessity followed by the removal of core in the presence of acidic condition (Fig 5)[143]. The formed nanocapsules serve as containers for drug, enzymes or other bioactive molecules. Since they are biodegradable they have an edge over other frequently used non-biodegradable templates (like polystyrene nanoparticles) as it has been reported that for some microcapsule systems core cannot be removed completely. These systems have also been used as template for the synthesis of porous silica nanoparticles[144] .

Ac ce p

888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905

te

d

M

869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887

Fig 5: Layer by layer deposition of oppositely charged polyelectrolyted on CaP/CaCO3nanoparticles followed by the core removal

5.8. Mineralization

Mineralization is deposition of minerals like calcium phosphate or calcium carbonate over other organic particles to develop robust and pH sensitive carrier systems. This kind of deposition of inorganic material over organic material has widened the choice of new functional nanoparticles. In the field of drug delivery stability of liposomes and micelles has always been a great concern. They suffer the limitation of low structural ability and therefore disassemble upon injection into blood stream and thus cannot be used for the preferential targeting the drug to a site or for sustained effect. Chemical cross linkers used to enhance their stability are mostly organic for which their toxicities are not well defined. Moreover the process of crosslinking requires complex chemistry and also acts as permanent barriers which reduce the drug release even at the target site. Earlier in inorganic deposition, silicification was the only choice to enhance the stability of micelles against disrupting chemical agents. 23 Page 24 of 47

an

us

cr

ip t

But recently CaP or CaCO3 has found to be better alternative showing superiority in respect of biocompatibility [145].Apart from enhancing the stability, coating of the CaP and CaCO3 over other nanoparticles also makes them to show pH dependent dissolution behavior and thus increasing their application in targeted or intracellular delivery without compromising the net payload (Fig 6). This kind of mineralization at the surface of organic nanoparticles does not change their structure since the process of deposition occurs without any chemical reaction. There are several publications reporting the deposition of CaP over micelles containing especially anticancer drugs loaded. It was observed that the release of doxorubicin from polymeric micelles of poly(ethylene glycol)-b-poly(l-aspartic acid)-b-poly(lphenylalanine) (PEG-PAsp-PPhe) was almost negligible in extracellular condition and significantly enhanced in the intracellular acidic conditions. Similarly, in another study release of doxorubicin was sustained at normal blood pH-7.4 but rate was much higher at mildly acidic conditions from mineralized PEGylated hyaluronic acid nanoparticles. The size of the mineralized particle (153.7 ± 4.5 nm) was also less than bare nanoparticles (265.1 ± 9.5 nm), which means that mineralization allows the formation of compact nanoparticles[146].

924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940

Ac ce p

te

d

M

906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923

Fig 6: Schematic representation of mineralization of nanoparticles showing sustained release at pH 7.4 but burst release at endosomal pH.

In another study mineralized chitosan-grafted-p(ethylene glycol)-dodecylamine (CMC-gPEG-DDA) micelles were developed which exhibited much enhanced serum stability as well pH dependant release at endosomal pH ( 3 PO< sub> 4 solution with Ca (OH)< sub> 2, Ultrasonics Sonochemistry, 8 (2001) 8588. [20] Z. Yang, Y. Huang, S.-T. Chen, Y.-Q. Zhao, H.-L. Li, Z.-A. Hu, Template synthesis of highly ordered hydroxyapatite nanowire arrays, Journal of materials science, 40 (2005) 1121-1125. [21] Q. He, Z. Huang, Y. Liu, W. Chen, T. Xu, Template-directed one-step synthesis of flowerlike porous carbonated hydroxyapatite spheres, Materials Letters, 61 (2007) 141-143. [22] J. Liu, K. Li, H. Wang, M. Zhu, H. Yan, Rapid formation of hydroxyapatite nanostructures by microwave irradiation, Chemical physics letters, 396 (2004) 429-432. [23] A. Escudero, M.E. Calvo, S. Rivera-Fernandez, J.M. de la Fuente, M. Ocana, Microwaveassisted synthesis of biocompatible europium-doped calcium hydroxyapatite and fluoroapatite luminescent nanospindles functionalized with poly(acrylic acid), Langmuir, 29 (2013) 1985-1994. [24] K. Lin, J. Chang, R. Cheng, M. Ruan, Hydrothermal microemulsion synthesis of stoichiometric single crystal hydroxyapatite nanorods with mono-dispersion and narrow-size distribution, Materials Letters, 61 (2007) 1683-1687. [25] J.-K. Han, H.-Y. Song, F. Saito, B.-T. Lee, Synthesis of high purity nano-sized hydroxyapatite powder by microwave-hydrothermal method, Materials chemistry and physics, 99 (2006) 235-239. [26] L.-y. Cao, C.-b. Zhang, J.-f. Huang, Synthesis of hydroxyapatite nanoparticles in ultrasonic precipitation, Ceramics International, 31 (2005) 1041-1044. [27] M. Fathi, E.M. Zahrani, Fabrication and characterization of fluoridated hydroxyapatite nanopowders via mechanical alloying, Journal of Alloys and Compounds, 475 (2009) 408414. [28] C.C. Silva, M.P.F. Graça, M.A. Valente, A.S.B. Sombra, Crystallite size study of nanocrystalline hydroxyapatite and ceramic system with titanium oxide obtained by dry ball milling, J Mater Sci, 42 (2007) 3851-3855. [29] X. Wang, J. Zhuang, Q. Peng, Y.D. Li, Liquid–Solid–Solution Synthesis of Biomedical Hydroxyapatite Nanorods, Advanced Materials, 18 (2006) 2031-2034.

34 Page 35 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[30] M. Boutinguiza, F. Lusquiños, A. Riveiro, R. Comesaña, J. Pou, Hydroxylapatite nanoparticles obtained by fiber laser-induced fracture, Applied Surface Science, 255 (2009) 5382-5385. [31] J.L. Xu, K.A. Khor, Z.L. Dong, Y.W. Gu, R. Kumar, P. Cheang, Preparation and characterization of nano-sized hydroxyapatite powders produced in a radio frequency (rf) thermal plasma, Materials Science and Engineering: A, 374 (2004) 101-108. [32] S. Barinov, E. Belonogov, V. Ievlev, A. Kostyuchenko, V. Putlyaev, Y.D. Tret’yakov, V. Smirnov, I. Fadeeva, Synthesis of dense nanocrystalline hydroxyapatite films, Doklady physical chemistry, Springer, 2007, pp. 15-18. [33] I. Roy, S. Mitra, A. Maitra, S. Mozumdar, Calcium phosphate nanoparticles as novel nonviral vectors for targeted gene delivery, International Journal of Pharmaceutics, 250 (2003) 25-33. [34] K. Kandori, T. Kuroda, S. Togashi, E. Katayama, Preparation of calcium hydroxyapatite nanoparticles using microreactor and their characteristics of protein adsorption, J Phys Chem B, 115 (2011) 653-659. [35] C. Lai, S. Tang, Y. Wang, K. Wei, Formation of calcium phosphate nanoparticles in reverse microemulsions, Materials Letters, 59 (2005) 210-214. [36] C. García, C. García, C. Paucar, Controlling morphology of hydroxyapatite nanoparticles through hydrothermal microemulsion chemical synthesis, Inorganic Chemistry Communications, 20 (2012) 90-92. [37] V. Uskokovic, D.P. Uskokovic, Nanosized hydroxyapatite and other calcium phosphates: chemistry of formation and application as drug and gene delivery agents, J Biomed Mater Res B Appl Biomater, 96 (2011) 152-191. [38] D.S.K. Bikramjit Basu, Ashok Kumar Advanced Biomaterials: Fundamentals, Processing, and Applications Wiley, 2009, pp. 776. [39] M.P. Ferraz, F.J. Monteiro, C.M. Manuel, Hydroxyapatite nanoparticles: A review of preparation methodologies, J Appl Biomater Biomech, 2 (2004) 74-80. [40] M. Nelson, G. Balasundaram, T.J. Webster, Increased osteoblast adhesion on nanoparticulate crystalline hydroxyapatite functionalized with KRSR, Int J Nanomedicine, 1 (2006) 339-349. [41] H. Wang, M. Bongio, K. Farbod, A.W. Nijhuis, J. van den Beucken, O.C. Boerman, J.C. van Hest, Y. Li, J.A. Jansen, S.C. Leeuwenburgh, Development of injectable organic/inorganic colloidal composite gels made of self-assembling gelatin nanospheres and calcium phosphate nanocrystals, Acta Biomater, 10 (2014) 508-519. [42] N. Thi Phuong, D. Bach Hai Phuong, D. Dinh Vu, N. Cuu Khoa, T. Ngoc Quyen, Enzymemediated in situ preparation of biocompatible hydrogel composites from chitosan derivative and biphasic calcium phosphate nanoparticles for bone regeneration, Advances in Natural Sciences: Nanoscience and Nanotechnology, 5 (2014) 015012. [43] D. Mohn, D. Ege, K. Feldman, O.D. Schneider, T. Imfeld, A.R. Boccaccini, W.J. Stark, Spherical calcium phosphate nanoparticle fillers allow polymer processing of bone fixation devices with high bioactivity, Polymer Engineering & Science, 50 (2010) 952-960. [44] M.P. Ginebra, C. Canal, M. Espanol, D. Pastorino, E.B. Montufar, Calcium phosphate cements as drug delivery materials, Adv Drug Deliv Rev, 64 (2012) 1090-1110. [45] M.D. Krebs, E. Salter, E. Chen, K.A. Sutter, E. Alsberg, Calcium phosphate-DNA nanoparticle gene delivery from alginate hydrogels induces in vivo osteogenesis, J Biomed Mater Res A, 92 (2010) 1131-1138.

35 Page 36 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[46] M. Bongio, J.J. van den Beucken, M.R. Nejadnik, Z. Tahmasebi Birgani, P. Habibovic, L.A. Kinard, F.K. Kasper, A.G. Mikos, S.C. Leeuwenburgh, J.A. Jansen, Subcutaneous tissue response and osteogenic performance of calcium phosphate nanoparticle-enriched hydrogels in the tibial medullary cavity of guinea pigs, Acta Biomater, 9 (2013) 5464-5474. [47] T.A. Desai, V. Uskokovic, Calcium phosphate nanoparticles: a future therapeutic platform for the treatment of osteomyelitis?, Ther Deliv, 4 (2013) 643-645. [48] F. Chen, H. Wan, T. Xia, X. Guo, H. Wang, Y. Liu, X. Li, Promoted regeneration of mature blood vessels by electrospun fibers with loaded multiple pDNA-calcium phosphate nanoparticles, Eur J Pharm Biopharm, 85 (2013) 699-710. [49] X. Zhang, A. Kovtun, C. Mendoza-Palomares, M. Oulad-Abdelghani, F. Fioretti, S. Rinckenbach, D. Mainard, M. Epple, N. Benkirane-Jessel, SiRNA-loaded multi-shell nanoparticles incorporated into a multilayered film as a reservoir for gene silencing, Biomaterials, 31 (2010) 6013-6018. [50] M.A. Melo, L. Cheng, M.D. Weir, R.C. Hsia, L.K. Rodrigues, H.H. Xu, Novel dental adhesive containing antibacterial agents and calcium phosphate nanoparticles, J Biomed Mater Res B Appl Biomater, 101 (2013) 620-629. [51] J.L. Moreau, L. Sun, L.C. Chow, H.H. Xu, Mechanical and acid neutralizing properties and bacteria inhibition of amorphous calcium phosphate dental nanocomposite, J Biomed Mater Res B Appl Biomater, 98 (2011) 80-88. [52] M.D. Weir, L.C. Chow, H.H. Xu, Remineralization of demineralized enamel via calcium phosphate nanocomposite, J Dent Res, 91 (2012) 979-984. [53] M.A. Melo, M.D. Weir, L.K. Rodrigues, H.H. Xu, Novel calcium phosphate nanocomposite with caries-inhibition in a human in situ model, Dent Mater, 29 (2013) 231240. [54] A. Kovtun, D. Kozlova, K. Ganesan, C. Biewald, N. Seipold, P. Gaengler, W.H. Arnold, M. Epple, Chlorhexidine-loaded calcium phosphate nanoparticles for dental maintenance treatment: combination of mineralising and antibacterial effects, RSC Advances, 2 (2012) 870-875. [55] M.A.S. Melo, L. Cheng, K. Zhang, M.D. Weir, L.K.A. Rodrigues, H.H.K. Xu, Novel dental adhesives containing nanoparticles of silver and amorphous calcium phosphate, Dental Materials, 29 (2013) 199-210. [56] V.C. Mendes, R. Moineddin, J.E. Davies, Discrete calcium phosphate nanocrystalline deposition enhances osteoconduction on titanium-based implant surfaces, Journal of Biomedical Materials Research Part A, 90A (2009) 577-585. [57] S. Yasuda, S. Kyotani, Nano-scaled hydroxyapatite/polymer composite IV. Fabrication and cell adhesion properties of a three-dimensional scaffold made of composite material with a silk fibroin substrate to develop a percutaneous device, Journal of Artificial Organs, 7 (2004) 137-144. [58] M. Okada, M. Masuda, R. Tanaka, K. Miyatake, D. Kuroda, T. Furuzono, Preparation of hydroxyapatite-nanocrystal-coated stainless steel, and its cell interaction, Journal of Biomedical Materials Research Part A, 86A (2008) 589-596. [59] H. Kadono, T. Furuzono, M. Masuda, M. Okada, M. Ueki, K. Takamizawa, R. Tanaka, K. Miyatake, Y. Koyama, K. Takakuda, In Vivo Evaluation of Hydroxyapatite Nanocoating on Polyester Artificial Vascular Grafts and Possibility as Soft-Tissue Compatible Material, ASAIO Journal, 56 (2010) 61-66 10.1097/MAT.1090b1013e3181c1945ae.

36 Page 37 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[60] H.S. Alghamdi, R. Bosco, S.K. Both, M. Iafisco, S.C. Leeuwenburgh, J.A. Jansen, J.J. van den Beucken, Synergistic effects of bisphosphonate and calcium phosphate nanoparticles on peri-implant bone responses in osteoporotic rats, Biomaterials, (2014). [61] M.A. Mintzer, E.E. Simanek, Nonviral vectors for gene delivery, Chem Rev, 109 (2009) 259-302. [62] J. Wang, J.-S. Chen, J.-Y. Zong, D. Zhao, F. Li, R.-X. Zhuo, S.-X. Cheng, Calcium Carbonate/Carboxymethyl Chitosan Hybrid Microspheres and Nanospheres for Drug Delivery, The Journal of Physical Chemistry C, 114 (2010) 18940-18945. [63] K. Roebrock, M. Wolf, S. Bovens, M. Lehr, C. Sunderkotter, Inhibition of benzalkonium chloride-induced skin inflammation in mice by an indol-1-ylpropan-2-one inhibitor of cytosolic phospholipase A2 alpha, Br J Dermatol, 166 (2012) 306-316. [64] L. Qin, Y. Sun, P. Liu, Q. Wang, B. Han, Y. Duan, F127/Calcium phosphate hybrid nanoparticles: a promising vector for improving siRNA delivery and gene silencing, J Biomater Sci Polym Ed, 24 (2013) 1757-1766. [65] C. Qi, Y.J. Zhu, F. Chen, Microwave hydrothermal transformation of amorphous calcium carbonate nanospheres and application in protein adsorption, ACS Appl Mater Interfaces, 6 (2014) 4310-4320. [66] E.V. Giger, J. Puigmartí-Luis, R. Schlatter, B. Castagner, P.S. Dittrich, J.-C. Leroux, Gene delivery with bisphosphonate-stabilized calcium phosphate nanoparticles, Journal of Controlled Release, 150 (2011) 87-93. [67] Y. Yang, J. Li, F. Liu, L. Huang, Systemic delivery of siRNA via LCP nanoparticle efficiently inhibits lung metastasis, Mol Ther, 20 (2012) 609-615. [68] C. Zhou, B. Yu, X. Yang, T. Huo, L.J. Lee, R.F. Barth, R.J. Lee, Lipid-coated nano-calciumphosphate (LNCP) for gene delivery, Int J Pharm, 392 (2010) 201-208. [69] Y. Kakizawa, S. Furukawa, K. Kataoka, Block copolymer-coated calcium phosphate nanoparticles sensing intracellular environment for oligodeoxynucleotide and siRNA delivery, J Control Release, 97 (2004) 345-356. [70] E.V. Giger, J. Puigmarti-Luis, R. Schlatter, B. Castagner, P.S. Dittrich, J.C. Leroux, Gene delivery with bisphosphonate-stabilized calcium phosphate nanoparticles, J Control Release, 150 (2011) 87-93. [71] K. Lee, M.H. Oh, M.S. Lee, Y.S. Nam, T.G. Park, J.H. Jeong, Stabilized calcium phosphate nano-aggregates using a dopa-chitosan conjugate for gene delivery, International Journal of Pharmaceutics, 445 (2013) 196-202. [72] A.S. Schenk, B. Cantaert, Y.-Y. Kim, Y. Li, E.S. Read, M. Semsarilar, S.P. Armes, F.C. Meldrum, Systematic Study of the Effects of Polyamines on Calcium Carbonate Precipitation, Chemistry of Materials, 26 (2014) 2703-2711. [73] C.Q. Wang, J.L. Wu, R.X. Zhuo, S.X. Cheng, Protamine sulfate-calcium carbonate-plasmid DNA ternary nanoparticles for efficient gene delivery, Mol Biosyst, 10 (2014) 672-678. [74] B. Mostaghaci, B. Loretz, R. Haberkorn, G. Kickelbick, C.-M. Lehr, One-Step Synthesis of Nanosized and Stable Amino-Functionalized Calcium Phosphate Particles for DNA Transfection, Chemistry of Materials, 25 (2013) 3667-3674. [75] Y. Sun, X. Li, X. Liang, Z. Wan, Y. Duan, Calcium Phosphate/Octadecyl-Quatemized Carboxymethyl Chitosan Nanoparticles: An Efficient and Promising Carrier for Gene Transfection, Journal of Nanoscience and Nanotechnology, 13 (2013) 5260-5266. [76] J. Klesing, S. Chernousova, M. Epple, Freeze-dried cationic calcium phosphate nanorods as versatile carriers of nucleic acids (DNA, siRNA), Journal of Materials Chemistry, 22 (2012) 199-204. 37 Page 38 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[77] V.V. Sokolova, I. Radtke, R. Heumann, M. Epple, Effective transfection of cells with multi-shell calcium phosphate-DNA nanoparticles, Biomaterials, 27 (2006) 3147-3153. [78] F. Pittella, H. Cabral, Y. Maeda, P. Mi, S. Watanabe, H. Takemoto, H.J. Kim, N. Nishiyama, K. Miyata, K. Kataoka, Systemic siRNA delivery to a spontaneous pancreatic tumor model in transgenic mice by PEGylated calcium phosphate hybrid micelles, Journal of Controlled Release, 178 (2014) 18-24. [79] S. Jang, S. Lee, H. Kim, J. Ham, J.-H. Seo, Y. Mok, M. Noh, Y. Lee, Preparation of pHsensitive CaP nanoparticles coated with a phosphate-based block copolymer for efficient gene delivery, Polymer, 53 (2012) 4678-4685. [80] T. Devarasu, R. Saad, A. Ouadi, B. Frisch, E. Robinet, P. Laquerriere, J.-C. Voegel, T. Baumert, J. Ogier, F. Meyer, Potent calcium phosphate nanoparticle surface coating for in vitro and in vivo siRNA delivery: a step toward multifunctional nanovectors, Journal of Materials Chemistry B, 1 (2013) 4692-4700. [81] D. Kozlova, S. Chernousova, T. Knuschke, J. Buer, A.M. Westendorf, M. Epple, Cell targeting by antibody-functionalized calcium phosphate nanoparticles, Journal of Materials Chemistry, 22 (2012) 396-404. [82] Y. Chen, L. Yang, S. Huang, Z. Li, L. Zhang, J. He, Z. Xu, L. Liu, Y. Cao, L. Sun, Delivery system for DNAzymes using arginine-modified hydroxyapatite nanoparticles for therapeutic application in a nasopharyngeal carcinoma model, Int J Nanomedicine, 8 (2013) 3107-3118. [83] S. Chen, D. Zhao, F. Li, R.-X. Zhuo, S.-X. Cheng, Co-delivery of genes and drugs with nanostructured calcium carbonate for cancer therapy, RSC Advances, 2 (2012) 1820-1826. [84] C.C. Tester, R.E. Brock, C.-H. Wu, M.R. Krejci, S. Weigand, D. Joester, In vitro synthesis and stabilization of amorphous calcium carbonate (ACC) nanoparticles within liposomes, CrystEngComm, 13 (2011) 3975-3978. [85] X.W. He, T. Liu, Y.X. Chen, D.J. Cheng, X.R. Li, Y. Xiao, Y.L. Feng, Calcium carbonate nanoparticle delivering vascular endothelial growth factor-C siRNA effectively inhibits lymphangiogenesis and growth of gastric cancer in vivo, Cancer Gene Ther, 15 (2008) 193202. [86] A.G.A. Coombes, S. Tasker, M. Lindblad, J. Holmgren, K. Hoste, V. Toncheva, E. Schacht, M.C. Davies, L. Illum, S.S. Davis, Biodegradable polymeric microparticles for drug delivery and vaccine formulation: the surface attachment of hydrophilic species using the concept of poly(ethylene glycol) anchoring segments, Biomaterials, 18 (1997) 1153-1161. [87] Q. He, A.R. Mitchell, S.L. Johnson, C. Wagner-Bartak, T. Morcol, S.J. Bell, Calcium phosphate nanoparticle adjuvant, Clin Diagn Lab Immunol, 7 (2000) 899-903. [88] Q. He, A. Mitchell, T. Morcol, S.J. Bell, Calcium phosphate nanoparticles induce mucosal immunity and protection against herpes simplex virus type 2, Clin Diagn Lab Immunol, 9 (2002) 1021-1024. [89] T. Knuschke, V. Sokolova, O. Rotan, M. Wadwa, M. Tenbusch, W. Hansen, P. Staeheli, M. Epple, J. Buer, A.M. Westendorf, Immunization with biodegradable nanoparticles efficiently induces cellular immunity and protects against influenza virus infection, J Immunol, 190 (2013) 6221-6229. [90] V. Sokolova, T. Knuschke, A. Kovtun, J. Buer, M. Epple, A.M. Westendorf, The use of calcium phosphate nanoparticles encapsulating Toll-like receptor ligands and the antigen hemagglutinin to induce dendritic cell maturation and T cell activation, Biomaterials, 31 (2010) 5627-5633. [91] V. Sokolova, T. Knuschke, J. Buer, A.M. Westendorf, M. Epple, Quantitative determination of the composition of multi-shell calcium phosphate-oligonucleotide 38 Page 39 of 47

Ac ce p

te

d

M

an

us

cr

ip t

nanoparticles and their application for the activation of dendritic cells, Acta Biomater, 7 (2011) 4029-4036. [92] V.V. Temchura, D. Kozlova, V. Sokolova, K. Uberla, M. Epple, Targeting and activation of antigen-specific B-cells by calcium phosphate nanoparticles loaded with protein antigen, Biomaterials, 35 (2014) 6098-6105. [93] D. Kozlova, V. Sokolova, M. Zhong, E. Zhang, J. Yang, W. Li, Y. Yang, J. Buer, A.M. Westendorf, M. Epple, H. Yan, Calcium phosphate nanoparticles show an effective activation of the innate immune response in vitro and in vivo after functionalization with flagellin, Virol Sin, 29 (2014) 33-39. [94] W. Zhou, A.O. Moguche, D. Chiu, K. Murali-Krishna, F. Baneyx, Just-in-time vaccines: Biomineralized calcium phosphate core-immunogen shell nanoparticles induce long-lasting CD8+ T cell responses in mice, Nanomedicine: Nanotechnology, Biology and Medicine, 10 (2014) 571-578. [95] A.K. Goyal, K. Khatri, N. Mishra, A. Mehta, B. Vaidya, S. Tiwari, R. Paliwal, S. Paliwal, S.P. Vyas, Development of self-assembled nanoceramic carrier construct(s) for vaccine delivery, J Biomater Appl, 24 (2009) 65-84. [96] T. Behera, P. Swain, Antigen adsorbed calcium phosphate nanoparticles stimulate both innate and adaptive immune response in fish, Labeo rohita H, Cell Immunol, 271 (2011) 350359. [97] K. Viswanathan, V.P. Gopinath, G.D. Raj, Formulation of Newcastle disease virus coupled calcium phosphate nanoparticles: An effective strategy for oculonasal delivery to chicken, Colloids Surf B Biointerfaces, 116 (2014) 9-16. [98] R. Ramachandran, W. Paul, C.P. Sharma, Synthesis and characterization of PEGylated calcium phosphate nanoparticles for oral insulin delivery, J Biomed Mater Res B Appl Biomater, 88 (2009) 41-48. [99] W. Paul, C.P. Sharma, Fatty Acid Conjugated Calcium Phosphate Nanoparticles for Protein Delivery, International Journal of Applied Ceramic Technology, 7 (2010) 129-138. [100] M. Higaki, M. Kameyama, M. Udagawa, Y. Ueno, Y. Yamaguchi, R. Igarashi, T. Ishihara, Y. Mizushima, Transdermal delivery of CaCO3-nanoparticles containing insulin, Diabetes Technol Ther, 8 (2006) 369-374. [101] S.K. Kim, M.B. Foote, L. Huang, Targeted delivery of EV peptide to tumor cell cytoplasm using lipid coated calcium carbonate nanoparticles, Cancer Lett, 334 (2013) 311318. [102] X. Cao, W. Deng, R. Qu, Q. Yu, J. Li, Y. Yang, Y. Cao, X. Gao, X. Xu, J. Yu, Non-Viral CoDelivery of the Four Yamanaka Factors for Generation of Human Induced Pluripotent Stem Cells via Calcium Phosphate Nanocomposite Particles, Advanced Functional Materials, 23 (2013) 5403-5411. [103] Y. Lv, H. Huang, B. Yang, H. Liu, Y. Li, J. Wang, A robust pH-sensitive drug carrier: Aqueous micelles mineralized by calcium phosphate based on chitosan, Carbohydrate Polymers, 111 (2014) 101-107. [104] N.P. Praetorius, T.K. Mandal, Engineered nanoparticles in cancer therapy, Recent Pat Drug Deliv Formul, 1 (2007) 37-51. [105] A. Kumar, G.K. Gupta, V. Khedgikar, J. Gautam, P. Kushwaha, B. Changkija, G.K. Nagar, V. Gupta, A. Verma, A.K. Dwivedi, N. Chattopadhyay, P.R. Mishra, R. Trivedi, In vivo efficacy studies of layer-by-layer nano-matrix bearing kaempferol for the conditions of osteoporosis: a study in ovariectomized rat model, Eur J Pharm Biopharm, 82 (2012) 508-517.

39 Page 40 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[106] G.K. Gupta, A. Kumar, V. Khedgikar, P. Kushwaha, J. Gautam, G.K. Nagar, V. Gupta, A. Verma, A.K. Dwivedi, A. Misra, R. Trivedi, P.R. Mishra, Osteogenic efficacy enhancement of kaempferol through an engineered layer-by-layer matrix: a study in ovariectomized rats, Nanomedicine, 8 (2013) 757-771. [107] X. Cheng, L. Kuhn, Chemotherapy drug delivery from calcium phosphate nanoparticles, Int J Nanomedicine, 2 (2007) 667-674. [108] A. Barroug, L.T. Kuhn, L.C. Gerstenfeld, M.J. Glimcher, Interactions of cisplatin with calcium phosphate nanoparticles: In vitro controlled adsorption and release, Journal of Orthopaedic Research, 22 (2004) 703-708. [109] U. Mukesh, V. Kulkarni, R. Tushar, R.S. Murthy, Methotrexate loaded self stabilized calcium phosphate nanoparticles: a novel inorganic carrier for intracellular drug delivery, J Biomed Nanotechnol, 5 (2009) 99-105. [110] Y. Luo, Y. Ling, W. Guo, J. Pang, W. Liu, Y. Fang, X. Wen, K. Wei, X. Gao, Docetaxel loaded oleic acid-coated hydroxyapatite nanoparticles enhance the docetaxel-induced apoptosis through activation of caspase-2 in androgen independent prostate cancer cells, J Control Release, 147 (2010) 278-288. [111] Y. Zhang, W.Y. Kim, L. Huang, Systemic delivery of gemcitabine triphosphate via LCP nanoparticles for NSCLC and pancreatic cancer therapy, Biomaterials, 34 (2013) 3447-3458. [112] Y. Luo, Y. Ling, W. Guo, J. Pang, W. Liu, Y. Fang, X. Wen, K. Wei, X. Gao, Docetaxel loaded oleic acid-coated hydroxyapatite nanoparticles enhance the docetaxel-induced apoptosis through activation of caspase-2 in androgen independent prostate cancer cells, Journal of controlled release : official journal of the Controlled Release Society, 147 (2010) 278-288. [113] X.Y. Zhao, Y.J. Zhu, F. Chen, B.Q. Lu, C. Qi, J. Zhao, J. Wu, Calcium phosphate hybrid nanoparticles: self-assembly formation, characterization, and application as an anticancer drug nanocarrier, Chem Asian J, 8 (2013) 1306-1312. [114] C. Yang, L. Han, F. Ma, L. Zhou, J. Zhou, S. Wei, G. Huang, J. Shen, F. Li, Synthesis of Calcium Phosphate Nanoparticle-Based Docetaxel Delivery System and its In Vitro Anticancer Activity, International Journal of Applied Ceramic Technology, (2013) n/a-n/a. [115] J. Yao, Y. Zhang, S. Ramishetti, Y. Wang, L. Huang, Turning an antiviral into an anticancer drug: Nanoparticle delivery of acyclovir monophosphate, Journal of Controlled Release, 170 (2013) 414-420. [116] M. Pourbaghi-Masouleh, V. Hosseini, Amorphous calcium phosphate nanoparticles could function as a novel cancer therapeutic agent by employing a suitable targeted drug delivery platform, Nanoscale Res Lett, 8 (2013) 1-6. [117] M. Horie, K. Nishio, H. Kato, S. Endoh, K. Fujita, A. Nakamura, S. Kinugasa, Y. Hagihara, Y. Yoshida, H. Iwahashi, Evaluation of cellular influences caused by calcium carbonate nanoparticles, Chem Biol Interact, 210 (2014) 64-76. [118] W. Tang, Y. Yuan, C. Liu, Y. Wu, X. Lu, J. Qian, Differential cytotoxicity and particle action of hydroxyapatite nanoparticles in human cancer cells, Nanomedicine, 9 (2013) 397412. [119] E.V. Shimanovskaia, O.V. Beznos, N.L. Kliachko, O.A. Kost, Nikol'skaia, II, T.A. Pavlenko, N.B. Chesnokova, A.V. Kabanov, [Production of timolol containing calcium-phosphate nanoparticles and evaluation of their effect on intraocular pressure in experiment], Vestn Oftalmol, 128 (2012) 15-18.

40 Page 41 of 47

cr

ip t

[120] T.C. Chu, Q. He, D.E. Potter, Biodegradable calcium phosphate nanoparticles as a new vehicle for delivery of a potential ocular hypotensive agent, Journal of ocular pharmacology and therapeutics, 18 (2002) 507-514. [121] R. Chen, Y. Qian, R. Li, Q. Zhang, D. Liu, M. Wang, Q. Xu, Methazolamide calcium phosphate nanoparticles in an ocular delivery system, Yakugaku Zasshi, 130 (2010) 419-424. [122] Y. Ueno, H. Futagawa, Y. Takagi, A. Ueno, Y. Mizushima, Drug-incorporating calcium carbonate nanoparticles for a new delivery system, Journal of controlled release, 103 (2005) 93-98. [123] S. Huang, J.C. Chen, C.W. Hsu, W.H. Chang, Effects of nano calcium carbonate and nano calcium citrate on toxicity in ICR mice and on bone mineral density in an ovariectomized mice model, Nanotechnology, 20 (2009) 0957-4484.

us

[124] F. Wang, Y. Zhang, X. Fan, M. Wang, Facile synthesis of water-soluble LaF3∶ Ln3+

Ac ce p

te

d

M

an

nanocrystals, Journal of Materials Chemistry, 16 (2006) 1031-1034. [125] J. Devoisselle, S. Soulie-Begu, S. Mordon, T. Desmettre, H. Maillols, A preliminary study of the in vivo behaviour of an emulsion formulation of indocyanine green, Lasers in medical science, 13 (1998) 279-282. [126] S.P. Mondejar, A. Kovtun, M. Epple, Lanthanide-doped calcium phosphate nanoparticles with high internal crystallinity and with a shell of DNA as fluorescent probes in cell experiments, Journal of Materials Chemistry, 17 (2007) 4153-4159. [127] M. Neumeier, L.A. Hails, S.A. Davis, S. Mann, M. Epple, Synthesis of fluorescent coreshell hydroxyapatite nanoparticles, Journal of Materials Chemistry, 21 (2011) 1250-1254. [128] E.I. Altinoglu, T.J. Russin, J.M. Kaiser, B.M. Barth, P.C. Eklund, M. Kester, J.H. Adair, Near-infrared emitting fluorophore-doped calcium phosphate nanoparticles for in vivo imaging of human breast cancer, ACS Nano, 2 (2008) 2075-2084. [129] K. Ganesan, A. Kovtun, S. Neumann, R. Heumann, M. Epple, Calcium phosphate nanoparticles: Colloidally stabilized and made fluorescent by a phosphate-functionalized porphyrin, J. Mater. Chem., 18 (2008) 3655-3661. [130] J. Klesing, A. Wiehe, B. Gitter, S. Grafe, M. Epple, Positively charged calcium phosphate/polymer nanoparticles for photodynamic therapy, J Mater Sci Mater Med, 21 (2010) 887-892. [131] J. Schwiertz, A. Wiehe, S. Gräfe, B. Gitter, M. Epple, Calcium phosphate nanoparticles as efficient carriers for photodynamic therapy against cells and bacteria, Biomaterials, 30 (2009) 3324-3331. [132] Y.C. Chen, A. Ostafin, H. Mizukami, Synthesis and characterization of pH sensitive carboxySNARF-1 nanoreactors, Nanotechnology, 21 (2010) 0957-4484. [133] P. Caravan, C.T. Farrar, L. Frullano, R. Uppal, Influence of molecular parameters and increasing magnetic field strength on relaxivity of gadolinium- and manganese-based T1 contrast agents, Contrast Media Mol Imaging, 4 (2009) 89-100. [134] Y.C. Tseng, Z. Xu, K. Guley, H. Yuan, L. Huang, Lipid-calcium phosphate nanoparticles for delivery to the lymphatic system and SPECT/CT imaging of lymph node metastases, Biomaterials, 35 (2014) 4688-4698. [135] A. Ashokan, G.S. Gowd, V.H. Somasundaram, A. Bhupathi, R. Peethambaran, A.K.K. Unni, S. Palaniswamy, S.V. Nair, M. Koyakutty, Multifunctional calcium phosphate nano41 Page 42 of 47

Ac ce p

te

d

M

an

us

cr

ip t

contrast agent for combined nuclear, magnetic and near-infrared in vivo imaging, Biomaterials, 34 (2013) 7143-7157. [136] D. Zhao, R.-X. Zhuo, S.-X. Cheng, Alginate modified nanostructured calcium carbonate with enhanced delivery efficiency for gene and drug delivery, Molecular BioSystems, 8 (2012) 753-759. [137] D. Zhao, R.X. Zhuo, S.X. Cheng, Modification of calcium carbonate based gene and drug delivery systems by a cell-penetrating peptide, Mol Biosyst, 8 (2012) 3288-3294. [138] J. Wang, B. Chen, D. Zhao, Y. Peng, R.X. Zhuo, S.X. Cheng, Peptide decorated calcium phosphate/carboxymethyl chitosan hybrid nanoparticles with improved drug delivery efficiency, Int J Pharm, 446 (2013) 205-210. [139] M. Kester, Y. Heakal, T. Fox, A. Sharma, G.P. Robertson, T.T. Morgan, E.I. Altinoglu, A. Tabakovic, M.R. Parette, S.M. Rouse, V. Ruiz-Velasco, J.H. Adair, Calcium phosphate nanocomposite particles for in vitro imaging and encapsulated chemotherapeutic drug delivery to cancer cells, Nano Lett, 8 (2008) 4116-4121. [140] T.T. Morgan, H.S. Muddana, E.I. Altinoglu, S.M. Rouse, A. Tabakovic, T. Tabouillot, T.J. Russin, S.S. Shanmugavelandy, P.J. Butler, P.C. Eklund, J.K. Yun, M. Kester, J.H. Adair, Encapsulation of organic molecules in calcium phosphate nanocomposite particles for intracellular imaging and drug delivery, Nano Lett, 8 (2008) 4108-4115. [141] F. Chen, P. Huang, Y.J. Zhu, J. Wu, D.X. Cui, Multifunctional Eu3+/Gd3+ dual-doped calcium phosphate vesicle-like nanospheres for sustained drug release and imaging, Biomaterials, 33 (2012) 6447-6455. [142] X.Y. Yang, Y.X. Li, M. Li, L. Zhang, L.X. Feng, N. Zhang, Hyaluronic acid-coated nanostructured lipid carriers for targeting paclitaxel to cancer, Cancer Lett, 334 (2013) 338345. [143] J. Schwiertz, W. Meyer-Zaika, L. Ruiz-Gonzalez, J.M. Gonzalez-Calbet, M. Vallet-Regi, M. Epple, Calcium phosphate nanoparticles as templates for nanocapsules prepared by the layer-by-layer technique, Journal of Materials Chemistry, 18 (2008) 3831-3834. [144] J.-F. Chen, H.-M. Ding, J.-X. Wang, L. Shao, Preparation and characterization of porous hollow silica nanoparticles for drug delivery application, Biomaterials, 25 (2004) 723-727. [145] H. Lee, S. Lee, Fabrication of core–shell hybrid nanoparticles by mineralization on poly(ε-caprolactone)-b-poly(methacrylic acid) copolymer micelles, Polymer Bulletin, 65 (2010) 743-752. [146] H.S. Han, J. Lee, H.R. Kim, S.Y. Chae, M. Kim, G. Saravanakumar, H.Y. Yoon, D.G. You, H. Ko, K. Kim, I.C. Kwon, J.C. Park, J.H. Park, Robust PEGylated hyaluronic acid nanoparticles as the carrier of doxorubicin: Mineralization and its effect on tumor targetability in vivo, Journal of Controlled Release, 168 (2013) 105-114. [147] Y. Lv, H. Huang, B. Yang, H. Liu, Y. Li, J. Wang, A robust pH-sensitive drug carrier: Aqueous micelles mineralized by calcium phosphate based on chitosan, Carbohydrate Polymers. [148] T. Ito, M. Takemasa, K. Makino, M. Otsuka, Preparation of calcium phosphate nanocapsules including simvastatin/deoxycholic acid assembly, and their therapeutic effect in osteoporosis model mice, Journal of Pharmacy and Pharmacology, 65 (2013) 494-502. [149] Z. Chen, Z. Li, Y. Lin, M. Yin, J. Ren, X. Qu, Biomineralization inspired surface engineering of nanocarriers for pH-responsive, targeted drug delivery, Biomaterials, 34 (2013) 1364-1371.

42 Page 43 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[150] H.P. Rim, K.H. Min, H.J. Lee, S.Y. Jeong, S.C. Lee, pH-Tunable Calcium Phosphate Covered Mesoporous Silica Nanocontainers for Intracellular Controlled Release of Guest Drugs, Angewandte Chemie International Edition, 50 (2011) 8853-8857. [151] M. Sivasubramanian, T. Thambi, J.H. Park, Mineralized cyclodextrin nanoparticles for sustained protein delivery, Carbohydrate Polymers, 97 (2013) 643-649. [152] B.P. Bastakoti, Y.-C. Hsu, S.-H. Liao, K.C.W. Wu, M. Inoue, S.-i. Yusa, K. Nakashima, Y. Yamauchi, Inorganic–Organic Hybrid Nanoparticles with Biocompatible Calcium Phosphate Thin Shells for Fluorescence Enhancement, Chemistry – An Asian Journal, 8 (2013) 13011305. [153] A. Sugawara, S. Yamane, K. Akiyoshi, Nanogel-Templated Mineralization: PolymerCalcium Phosphate Hybrid Nanomaterials, Macromolecular Rapid Communications, 27 (2006) 441-446. [154] E.-J. Cha, I.-C. Sun, S. Lee, K. Kim, I. Kwon, C.-H. Ahn, Development of a pH sensitive nanocarrier using calcium phosphate coated gold nanoparticles as a platform for a potential theranostic material, Macromol. Res., 20 (2012) 319-326. [155] Z. Tang, Y. Zhou, H. Sun, D. Li, S. Zhou, Biodegradable magnetic calcium phosphate nanoformulation for cancer therapy, Eur J Pharm Biopharm, (2014). [156] W.M. Li, S.Y. Chen, D.M. Liu, In situ doxorubicin-CaP shell formation on amphiphilic gelatin-iron oxide core as a multifunctional drug delivery system with improved cytocompatibility, pH-responsive drug release and MR imaging, Acta Biomater, 9 (2013) 5360-5368. [157] K. Scholler, A. Ethirajan, A. Zeller, K. Landfester, Biomimetic Route to Calcium Phosphate Coated Polymeric Nanoparticles: Influence of Different Functional Groups and pH, Macromolecular Chemistry and Physics, 212 (2011) 1165-1175. [158] K.K. Perkin, J.L. Turner, K.L. Wooley, S. Mann, Fabrication of hybrid nanocapsules by calcium phosphate mineralization of shell cross-linked polymer micelles and nanocages, Nano Lett, 5 (2005) 1457-1461. [159] Q. Xu, Y. Tanaka, J.T. Czernuszka, Encapsulation and release of a hydrophobic drug from hydroxyapatite coated liposomes, Biomaterials, 28 (2007) 2687-2694. [160] D.Y. CHEN ChangJing, YAN ErYun, HU Yong, JIANG XiQun, Preparation of porous chitosan-poly(acrylic acid)- calcium phosphate hybrid nanoparticles via mineralization, Chinese Science Bulletin, 54 (2009) 3127-3136. [161] A. Hanifi, M.H. Fathi, H. Mir Mohammad Sadeghi, Effect of strontium ions substitution on gene delivery related properties of calcium phosphate nanoparticles, J Mater Sci Mater Med, 21 (2010) 2601-2609. [162] A. Peetsch, C. Greulich, D. Braun, C. Stroetges, H. Rehage, B. Siebers, M. Koller, M. Epple, Silver-doped calcium phosphate nanoparticles: synthesis, characterization, and toxic effects toward mammalian and prokaryotic cells, Colloids Surf B Biointerfaces, 102 (2013) 724-729. [163] J.H. Swet, H.J. Pacheco, D.A. Iannitti, A. El-Ghanam, I.H. McKillop, A silica-calciumphosphate nanocomposite drug delivery system for the treatment of hepatocellular carcinoma: In vivo study, Journal of Biomedical Materials Research Part B: Applied Biomaterials, 102 (2014) 190-202. [164] J. Kang, J. Yang, J. Lee, S.J. Oh, S. Moon, H.J. Lee, S.C. Lee, J.-H. Son, D. Kim, K. Lee, J.-S. Suh, Y.-M. Huh, S. Haam, Gold-layered calcium phosphate plasmonic resonants for localized photothermal treatment of human epithelial cancer, Journal of Materials Chemistry, 19 (2009) 2902-2905. 43 Page 44 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[165] Y. Cai, H. Pan, X. Xu, Q. Hu, L. Li, R. Tang, Ultrasonic controlled morphology transformation of hollow calcium phosphate nanospheres: a smart and biocompatible drug release system, Chemistry of materials, 19 (2007) 3081-3083. [166] K. Kawai, B.J. Larson, H. Ishise, A.L. Carre, S. Nishimoto, M. Longaker, H.P. Lorenz, Calcium-based nanoparticles accelerate skin wound healing, PloS one, 6 (2011) e27106. [167] S. Fujii, M. Okada, T. Nishimura, H. Maeda, T. Sugimoto, H. Hamasaki, T. Furuzono, Y. Nakamura, Hydroxyapatite-armored poly(ε-caprolactone) microspheres and hydroxyapatite microcapsules fabricated via a Pickering emulsion route, Journal of Colloid and Interface Science, 374 (2012) 1-8. [168] K.S. Soppimath, T.M. Aminabhavi, A.R. Kulkarni, W.E. Rudzinski, Biodegradable polymeric nanoparticles as drug delivery devices, J Control Release, 70 (2001) 1-20. [169] F. Qing, Z. Wang, Y. Hong, M. Liu, B. Guo, H. Luo, X. Zhang, Selective effects of hydroxyapatite nanoparticles on osteosarcoma cells and osteoblasts, J Mater Sci Mater Med, 23 (2012) 2245-2251. [170] S. Bisht, G. Bhakta, S. Mitra, A. Maitra, pDNA loaded calcium phosphate nanoparticles: highly efficient non-viral vector for gene delivery, International Journal of Pharmaceutics, 288 (2005) 157-168. [171] E.V. Giger, B. Castagner, J. Räikkönen, J. Mönkkönen, J.-C. Leroux, siRNA Transfection with Calcium Phosphate Nanoparticles Stabilized with PEGylated Chelators, Advanced Healthcare Materials, 2 (2013) 134-144. [172] J. Li, Y.-C. Chen, Y.-C. Tseng, S. Mozumdar, L. Huang, Biodegradable calcium phosphate nanoparticle with lipid coating for systemic siRNA delivery, Journal of Controlled Release, 142 (2010) 416-421. [173] C. Zhou, B. Yu, X. Yang, T. Huo, L.J. Lee, R.F. Barth, R.J. Lee, Lipid-coated nano-calciumphosphate (LNCP) for gene delivery, International Journal of Pharmaceutics, 392 (2010) 201208. [174] F. Pittella, H. Cabral, Y. Maeda, P. Mi, S. Watanabe, H. Takemoto, H.J. Kim, N. Nishiyama, K. Miyata, K. Kataoka, Systemic siRNA delivery to a spontaneous pancreatic tumor model in transgenic mice by PEGylated calcium phosphate hybrid micelles, J Control Release, 178 (2014) 18-24. [175] G.D. Venkatasubbu, S. Ramasamy, G.S. Avadhani, V. Ramakrishnan, J. Kumar, Surface modification and paclitaxel drug delivery of folic acid modified polyethylene glycol functionalized hydroxyapatite nanoparticles, Powder Technology, 235 (2013) 437-442. [176] K. Watanabe, Y. Nishio, R. Makiura, A. Nakahira, C. Kojima, Paclitaxel-loaded hydroxyapatite/collagen hybrid gels as drug delivery systems for metastatic cancer cells, Int J Pharm, 446 (2013) 81-86. [177] G.D. Venkatasubbu, S. Ramasamy, G.P. Reddy, J. Kumar, In vitro and In vivo anticancer activity of surface modified paclitaxel attached hydroxyapatite and titanium dioxide nanoparticles, Biomed Microdevices, 15 (2013) 711-726. [178] J. Yao, Y. Zhang, S. Ramishetti, Y. Wang, L. Huang, Turning an antiviral into an anticancer drug: nanoparticle delivery of acyclovir monophosphate, J Control Release, 170 (2013) 414-420. [179] B. Sarmento, A. Ribeiro, F. Veiga, P. Sampaio, R. Neufeld, D. Ferreira, Alginate/chitosan nanoparticles are effective for oral insulin delivery, Pharm Res, 24 (2007) 2198-2206. [180] W. Zhou, A.O. Moguche, D. Chiu, K. Murali-Krishna, F. Baneyx, Just-in-time vaccines: Biomineralized calcium phosphate core-immunogen shell nanoparticles induce long-lasting CD8(+) T cell responses in mice, Nanomedicine, 10 (2014) 571-578. 44 Page 45 of 47

Ac ce p

te

d

M

an

us

cr

ip t

[181] V. Sokolova, D. Kozlova, T. Knuschke, J. Buer, A.M. Westendorf, M. Epple, Mechanism of the uptake of cationic and anionic calcium phosphate nanoparticles by cells, Acta Biomaterialia, 9 (2013) 7527-7535. [182] A. Ashokan, G.S. Gowd, V.H. Somasundaram, A. Bhupathi, R. Peethambaran, A.K. Unni, S. Palaniswamy, S.V. Nair, M. Koyakutty, Multifunctional calcium phosphate nano-contrast agent for combined nuclear, magnetic and near-infrared in vivo imaging, Biomaterials, 34 (2013) 7143-7157. [183] Y. Zhang, L. Peng, R.J. Mumper, L. Huang, Combinational delivery of c-myc siRNA and nucleoside analogs in a single, synthetic nanocarrier for targeted cancer therapy, Biomaterials, 34 (2013) 8459-8468. [184] K.H. Min, H.J. Lee, K. Kim, I.C. Kwon, S.Y. Jeong, S.C. Lee, The tumor accumulation and therapeutic efficacy of doxorubicin carried in calcium phosphate-reinforced polymer nanoparticles, Biomaterials, 33 (2012) 5788-5797. [185] Q. Xu, Y. Tanaka, J.T. Czernuszka, Encapsulation and release of a hydrophobic drug from hydroxyapatite coated liposomes, Biomaterials, 28 (2007) 2687-2694.

45 Page 46 of 47

Ac ce p

te

d

M

an

us

cr

ip t

Graphical abstract

46 Page 47 of 47

An insight into functionalized calcium based inorganic nanomaterials in biomedicine: Trends and transitions.

Over the recent years the use of biocompatible and biodegradable nanoparticles in biomedicine has become a significant priority. Calcium based ceramic...
829KB Sizes 6 Downloads 13 Views