Научная статья на тему 'CaCO3 vaterites as components of target drug delivery systems'

CaCO3 vaterites as components of target drug delivery systems Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
224
35
i Надоели баннеры? Вы всегда можете отключить рекламу.
Журнал
Cellular Therapy and Transplantation
Scopus
ВАК
Ключевые слова
ANTICANCER DRUGS DELIVERY SYSTEMS / САСО3 VATERITES / BLOOD PLASMA / INTRAMUSCULAR ADMINISTRATION / BIORESORPTION

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — Sudareva Natalia N., Popryadukhin Pavel V., Saprykina Natalia N., Suvorova Olga M., Yukina Galina Yu.

Successful treatment of the majority of oncological diseases that affect solid organs is related to appropriate use of potent and (to varying degrees) toxic antitumor drugs. In a number of cases, chemotherapy requires the maximum localized action of a drug in the tumor area. The most efficient methods of drug administration are introducing medicinal compounds (MC) directly into the tumor or use of target drug delivery systems. The second method makes it possible to decrease general toxicity of MC, and to reach prolonged therapeutic action due to uniform and time-controlled release of a MC into tumor tissue. In the present work, we studied behavior of porous spherical СаСО3 vaterites (components of delivery systems for antitumor drugs) in various environments (human blood plasma, rat muscle tissue). It was demonstrated that the studied drug carriers undergo morphological transformations and are destructed with time. In blood plasma, due to ion exchange reactions, vaterites are transformed into gradually disintegrating needle-like structures (as shown by scanning electron microscopy and energy dispersive spectroscopy). Similar processes were observed in muscle tissue: in three days, spheres were transformed into needle-like structures and then underwent complete bioresorption.

i Надоели баннеры? Вы всегда можете отключить рекламу.
iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.
i Надоели баннеры? Вы всегда можете отключить рекламу.

Текст научной работы на тему «CaCO3 vaterites as components of target drug delivery systems»

Cellular Therapy and Transplantation (CTT). Vol. 9, No. 2, 2020 doi: 10.1862()/ctt-1866-8836-202()-9-2-13-19 Submitted: 13 May 2020, accepted: 06 June 2020

CaCO3 vaterites as components of target drug delivery systems

Natalia N. Sudareva 1,2> Pavel V. Popryadukhin 1,2> Natalia N. Saprykina Olga M. Suvorova Galina Yu. Yukina 2, Oleg V. Galibin 2, Aleksandr D. Vilesov 1,2

1 Institute of Macromolecular Compounds RAS, St. Petersburg, Russia

2 Pavlov University, St. Petersburg, Russia

Dr. Natalia N. Sudareva, Research Institute of Macromole- E-mail: nnsas@mail.ru

cular Compounds, Bolshoi Prosp. 31 (V.O.), 199004, St. Petersburg, Russia

Citation: Sudareva NN, Popryadukhin PV, Saprykina NN et al. CaCO3 vaterites as components of target drug delivery systems. Cell Ther Transplant 2020; 9(2): 13-19.

Summary

Successful treatment of the majority of oncological diseases that affect solid organs is related to appropriate use of potent and (to varying degrees) toxic antitumor drugs. In a number of cases, chemotherapy requires the maximum localized action of a drug in the tumor area. The most efficient methods of drug administration are introducing medicinal compounds (MC) directly into the tumor or use of target drug delivery systems. The second method makes it possible to decrease general toxicity of MC, and to reach prolonged therapeutic action due to uniform and time-controlled release of a MC into tumor tissue.

In the present work, we studied behavior of porous spherical СаСО3 vaterites (components of delivery systems for antitumor drugs) in various environments (human blood plasma, rat muscle tissue). It was demonstrated that the studied drug carriers undergo morphological transformations and are destructed with time.

In blood plasma, due to ion exchange reactions, vaterites are transformed into gradually disintegrating needle-like structures (as shown by scanning electron microscopy and energy dispersive spectroscopy). Similar processes were observed in muscle tissue: in three days, spheres were transformed into needle-like structures and then underwent complete bioresorption.

Keywords

Anticancer drugs delivery systems, СаСО3 vaterites, blood plasma, intramuscular administration, bioresorption.

Introduction

Target drug delivery systems find increasingly wide application in medicine. Use of these systems requires high stability of encapsulated MC, low dosage and toxicity, prolonged therapeutic action. Porous vaterites (one of three calcium carbonate polymorphs) have been used as carriers in delivery systems (DS) for biologically active compounds and medicinal compounds since 2004 [1]. In many research works, they were used as "sacrificed" matrices. Porous carbonate cores were saturated with biologically active compounds using different methods, then their surface was coated layer-by-layer with polyelectrolytes; polymers with opposite excess charges

were applied by turns. After dissolution of CaCO3 cores in the presence of chelate compounds (e.g., ethylenediamine-tetraacetic acid), these multilayered shells were used as capsules for delivery of biologically active compounds [2]. In some cases, carbonate cores were not dissolved, but used together with their PE shells [3-5]. Since one of the objectives of employing delivery systems is to provide prolonged release of an encapsulated MC, preservation of the porous core increases resistance of the structure against external influence and thus helps attain this goal. Another way of using CaCO3 as a component of DS consists in including carbonate cores into alginate granule, which significantly simplifies DS preparation [6].

A number of research papers [7-9] report preparation of DS with CaCO3 in combination with various polymers; antitumor drug doxorubicin was used as an active substance. In vitro experiments demonstrated prolonged pH-dependent release of the drug.

Note that synthesis of CaCO3 cores is relatively simple. It is believed that they are completely biocompatible and biodegradable; they show neither toxicity nor immunogenic-ity, and thus are well tolerated by a recipient organism [10]. This opinion was confirmed by the studies of behavior of CaCO3-based delivery systems in various model environments as well as upon administration of these DS into living rabbits, rats and mice by various methods. Configurations of DS based on CaCO3 cores depend on the method used for their administration. The influence of various environments on the DS containing CaCO3 cores is described in the papers that are quoted below.

In water or physiological solution (0.9% NaCl), CaCO3 vater-ites undergo morphological transformations [11]. At medium temperatures, porous vaterites turn into calcites (which are more thermodynamically stable), and at elevated temperatures (above 37-40°C), they are transformed into arago-nites [12]. Since these polymorphs are not porous, recrystal-lization is accompanied by release of drugs encapsulated in vaterites. The drug release profiles correlate with percentages of calcites formed [13].

Oral administration is the most convenient method for patients. However, vaterite cores dissolve in acidic medium of a stomach; therefore, the cores with encapsulated MC should be protected. This protection can be provided both by PE shells (on condition that their components are stable in acidic stomach environment) and alginate granules surrounding CaCO3 cores. Since it is necessary to provide penetration of MC from intestinal tract into main blood flow, a polymer shell should swell or dissolve in the middle division of intestinal tract, thus releasing CaCO3 with MC. Model experiments involving 0.15 M phosphate buffer with pH=7.4 (model intestinal fluid) demonstrated that CaCO3 enters into ion-exchange reaction with phosphate ions; as a result, rather compact porous vaterites are transformed into loose macroporous CaHP04 structures. This process facilitates release of the encapsulated MC. Scanning electron microscopy (SEM) and energy-dispersive spectroscopy (EDS) studies revealed structural changes in CaCO3 vaterites [14]. Similar transformation also occurs with time in the case of two-level DS that consist of alginate granules and carbonate cores. The presence of fragments of CaCO3 cores in blood and plasma of experimental animals (rats) was confirmed by elemental analysis of the samples [15].

The requirements for size of DS intended for parentheral administration are more rigid, but in this case the carrier should not be necessary protected (unlike the systems used in oral delivery). The CaCO3 vaterites that were synthesized according to the technique described in [1] have sizes of 3-5 ^m. Diameter of cores may be reduced by various methods: change in the basic synthesis conditions - increasing concentration of the initial solutions of salts (Na2CO3 and CaCl2), and stirring intensity [16]; increase in viscosity of starting solutions by adding ethylene glycol [17]; addition of

a polyelectrolyte during co-precipitation of the salts [18-20]. Unfortunately, the latter method gives low yield of the final product and requires monitoring interaction between MC and polymer.

The authors of [21] used intratracheal administration of CaCO3 cores that contained BSA protein labeled with Cy7 fluorophore. It was demonstrated that efficiency of penetration into lungs for carriers of various diameters decreased with increasing core size from 0.65 to 3.15 ^m. Penetration of the labeled protein into lungs with the aid of CaCO3 vaterites of different sizes was confirmed by confocal microscopy of mouse lung cryocut sections. The lower DS size, the deeper they penetrate into lung tissue. Confocal microscopy makes it possible to localize CaCO3 carriers in a sample. Recrystal-lization of vaterites was observed in the model environment that included physiological solution and bronchoalveolar lavage (containing proteins and surfactants). It was shown that the components of lavage covering vaterite surface protect them from recrystallization.

The authors of [22] demonstrated possibility of penetration of CaCO3 vaterites with encapsulated loperamide through blood-brain barrier of rats after intranasal administration. In order to enhance mucoadhesion, CaCO3 cores were covered with mucoadhesive polymers (hyaluronic acid or poly-L-lysine).

It was suggested [10] to use CaCO3 cores with encapsulated superoxide dismutase enzyme as an ophthalmic delivery system. According to the authors, no undesirable effects were observed after injections of vaterite microcrystals (concentration 10 mg/mL) into eye tissues of rabbits.

In vivo transdermal administration of CaCO3 particles (diameter: 4 ^m) to a depth of 200 ^m was performed via laser ablation followed by massage. These relatively large particles did not penetrate into the underlying derma. In 1 week after beginning of the experiment, CaCO3 particles dissolved in rat body and released the encapsulated compound [23].

It was revealed [24] that porous CaC03 cores degraded completely in three months after introducing them into rat bone tissue.

Along with other calcium-containing inorganic nanostruc-tured materials, CaCO3 vaterites find increasing applications in regenerative medicine and tissue engineering [25].

To summarize, all methods for introducing DS based on CaCO3 vaterites are aimed at providing absorption of cores by cells. The influence of size and shape of CaCO3 particles on cell uptake was studied in [26]. It was demonstrated that internalization is more effective for spherical particles with the lowest volume, and for elongated particles.

Currently, there are no literature data on the studies of behavior of vaterite-based DS in human blood plasma and upon their intramuscular administration. When using the majority of the above-mentioned methods, it is necessary to study transformations of DS in blood plasma. The second method may be efficient when DS with MC are introduced directly into tumor tissue. Thus, the goal of the present work was to study behavior of spherical CaCO3 vaterites (compo-

nents of target delivery systems for antitumor drugs) in vitro (in human blood plasma) and in vivo (in rat muscle tissue).

Abbreviations: DS, delivery systems; MC, medicinal compounds; EDS, energy dispersive spectroscopy; SEM, scanning electron microscopy; EDTA, ethylenediaminetetraacetic acid; BSA, bovine serum albumin.

Materials and methods

Synthesis of carbonate cores

Porous vaterites (СаСО3 cores) were prepared by co-precipitation according to the technique described in [1] with some modifications. Equal volumes of 1 M aqueous solutions of CaCl2x2H2O and Na2CO3 were rapidly mixed at stirring with an RW 20 anchor-type mechanical stirrer (Kika-Werk, Switzerland) (1000 rpm). The mixture was stirred for 30 s. Then the suspension was filtered through Schott filter glass (#16), washed thrice with distilled water, then with acetone/water mixtures with increasing acetone concentrations (33%, 50%, and 100%). The precipitate was dried in thermostat at 40-50°C until a constant weight was achieved. Diameter of the obtained cores varies from 1 to 3 ^m.

Interaction between СаСО3 and human blood plasma

Interaction between carbonate cores and human blood plasma was performed at continuous stirring of the suspension. When the reaction was complete, the cores were centrifuged (5 min at 3000 rpm); the supernatant was poured out and substituted for distilled water. The procedure was performed twice. The cores were dried at 40°C until a constant weight was achieved.

Scanning electron microscopy (SEM)

SEM microphotographs of СаСО3 cores were obtained with the help of a Supra 55VP scanning electron microscope (Carl Zeiss, Germany) using secondary electron imaging; before the experiments, the samples were coated with thin platinum layer.

Energy dispersive spectroscopy (EDS)

Elemental compositions of the samples were determined by energy-dispersive spectroscopy (EDS) using an X-Max 80 detector (Oxford Instruments, UK).

Experiments with animals

The experiments involving animals were performed according to the laboratory animal welfare policy accepted in Russian Federation and European Convention for the Protection of Vertebrate Animals used for Experimental and other Scientific Purposes (ETS 123, Strasbourg, 1986).

In vivo experiments involved 10 male Wistar rats (weight: 200-250 g, age: 3 months). Before studies of bioresorption in vivo, СаСО3 cores were sterilized in autoclave at 110°С for 1 h. Each weighed amount of СаСО3 (10 mg) intended for an experiment in each of two locations in one animal was carefully hermetically packed in aluminum foil. The animals were operated under general anesthesia (intra-peritoneal injections of Zoletil 100 (0.1 mL) and Rometar

(20 mg/mL) solutions, 0.0125 mL per 0.1 kg of animal body mass). The samples were placed into thigh great adductor muscles (musculus adductor magnus) of both hind extremities. Then the wounds were sutured layer by layer using atraumatic needles and Prolene 4-0 suture. After outer suturing, the rats were caged individually, were fed standard diet, and had free access to water. All animals were active after surgery; no inflammation in the implantation area was observed, which is indicative of the absence of detrimental effects of implantation.

Histological studies

In 1 and 2 weeks after operation, samples of muscle tissue containing СаСО3 were fixed with 10% neutral formalin in phosphate buffer (рН=7.4) for not less than 24 hrs, dehydrated using a series of ethanol solutions with increasing concentrations, and enclosed in paraffin blocks according to the standard histological technique. The paraffin cuts (5 ^m in width) transverse to muscular fibers were obtained with the use of an Accu-Cut SRT 200 microtome (Sakura, Japan) and stained with Mayer hematoxylin and eosin (Bio-Optica, Italy). The connective tissue was visualized according to the Mallory method (BioVitrum, Russia). Microscopic analysis was performed using a Leica DM750 light microscope (Germany) with a 10x ocular and 4, 10, 40, and 100x objectives. Images were recorded with an ICC50 camera (Leica, Germany).

Results

Influence of human blood plasma on the structure of СаСО3 cores

Blood plasma contains phosphate ions, which enter into reaction with CaCO3 vaterites; as a result, macroporous СаНРО4 structures are gradually formed [14]. It is seen in the SEM images of CaCO3 vaterites (Fig. 1) that the objects with increasingly loose structure are formed with time; they consist of needle-like subunits less than 1 ^m in diameter.

Phosphorus content in the studied structures was determined by energy-dispersive spectroscopy (see Table 1).

The EDS data show that phosphorus content in transformed structures increases with time; this result confirms that ion exchange reaction indeed occurs in CaCO3 vaterites.

Table 1. Phosphorus content (P) in CaCO3 samples that contacted with blood plasma for various periods of time

Sample Р (wt. %)

Plasma 0.10±0.07

СаСО3 2 hours in plasma 0.30±0.11

СаСО3 24 hours in plasma 0.38±0.16

СаСО3 50 hours in plasma 1.92±0.50

Figure 1. Microphotographs of CaCO3 vaterites taken upon interaction with human blood plasma for various periods of time: A - 2 hrs; B - 24 hrs; C - 50 hrs

Transformation of Ca^ vaterites upon intramuscular administration

After injection of CaCO3 vaterites into thigh great adductor muscles (musculus adductor magnus) of both hind extremities in rats, needle-like structures were formed (Fig. 2) and then gradually disappeared in two weeks due to bioresorption. Presumably, these needles are aragonites (one of three CaCO3 polymorphs). Fig. 2B presents the magnified image of the area where vaterites were introduced and then transformed into aragonites (1 week after operation). As was mentioned in Introduction, aragonites (non-porous elongated structures) are one of three morphological modifications of calcium carbonate, along with non-porous (usually cubic) calcites and porous spherical vaterites (which are used as components of target drug delivery systems). Transforma-

tion of vaterites during their use in delivery systems into cal-cites is frequently observed [13]. Formation of aragonite-like structures in the process of bioresorption of CaCO3 vaterites was revealed in the present work for the first time.

Discussion

The reason for transformation of porous СаСОз vaterites (diameter: 1 - 3 [im) into needle-like aragonites (length: 30 -150 [im, width: 10 - 40 [im) in muscle tissue still remains unclear. It may be suggested that morphological transformation of vaterites is influenced by the following factors. First, there is a difference between pH values of muscle tissue and blood or its components (pH of muscle tissue is lower). The second factor involves peculiarities of metabolic processes, mainly, exchange of carbon dioxide. Upon interaction with water,

Figure 2. Histological cuts of rat muscle tissue obtained in 1 week after implantation of CaCO3 vaterites. Staining with hematoxylin and eosin; objectives 10* (a), 40* (b)

carbon dioxide forms carbonic acid, which reacts with calcium carbonate. Among other factors are intensive action of immune cells, and, finally, mechanic action related to muscle contraction. This issue should be investigated further.

The comparison between our results and the literature data on transformation of СаСО3 vaterites with encapsulated Fe3O4 nanoparticles (which occurred after shallow transdermal injection into rat body [23]) shows that no vaterite modification in muscle tissue was observed. The histological sections prepared in one week after transdermal administration show spherical structures almost similar to the initial cores. In two weeks after operation, vaterites underwent bioresorption, and Fe3O4 nanoparticles were released. These data may indirectly confirm our hypothesis concerning the influence of the above factors on transformation of CaCO3 vaterites in muscle tissue.

Bioresorption of vaterites in blood plasma in vitro is also completed in relatively short period of time (several weeks), while plasma composition remains mostly unchanged.

The main advantage of the DS based on CaCO3 vaterites intended for intramuscular administration of antitumor preparations is the fact that modified carbonate cores undergo complete bioresorption in 2 weeks in vivo and exert no negative influence on the surrounding tissues. The fact that aragonites are formed in the muscles once again indicates the ambiguity of applying the conclusions obtained from in vitro experiments to the in vivo behavior of the studied objects.

The obtained results confirm ability of porous calcium carbonate cores for bioresorption and their safety for medicinal use, which allows us to recommend porous CaCO3 vaterites for further experimental studies as components of target drug delivery systems.

Conflict of interests

None declared.

References

1. Volodkin DV, Petrov AI, Prevot M, Sukhorukov GB. Matrix polyelectrolyte microcapsules: new system for mac-romolecule encapsulation. Langmuir. 2004; 20: 3398-3406.

2. She Z, Wang CX, Li J, Sukhorukov GB, Antipina MN. Encapsulation of basic fibroblast growth factor by polyelec-trolyte multilayer microcapsules and its controlled release for enhancing cell proliferation. Biomacromolecules. 2012; 13(7): 2174-2180. DOI: 10.1021/bm3005879.

3. Liu D, Jiang G, Yu W, Tong Z, Kong X, Yao J. Oral delivery of insulin using CaCO3-based composite nanocarriers with hyaluronic acid coatings. Materials Letters. 2017; 188: 263-266. DOI: 10.1016/j.matlet.2016.10.117.

4. Ramalapaa B, Crasson O, Vandevenne M, Cordonnier T, Galleni M, Boury F. Protein-polysaccharide complexes for enhanced protein delivery in hyaluronic acid templated calcium carbonate microparticles. J Mater Chem B. 2017; 5: 7360-7368. DOI: 10.1039/C7TB01538K.

5. Peng C, Zhao Q, Gao C. Sustained delivery of doxorubicin by porous CaCO3 and chitosan/alginate multilayers-coated CaCO3 microparticles. Colloids and Surfaces A: Physico-chem Eng Aspects. 2010; 353:132-139. D01:10.1016/j.col-surfa.2009.11.004.

6. Sudareva N, Suvorova O, Saprykina N, Vilesov A, Bel'tyu-kov P, Petunov S. Alginate-containing systems for oral delivery of superoxide dismutase. Comparison of various configurations and their properties. J Microencapsulation. 2016; 33(5): 487-496. DOI: 10.1080/02652048.2016.1206146.

7. Zhao D, Zhuo R, Cheng S. Alginate modified nanostruc-tured calcium carbonate with enhanced delivery efficiency for gene and drug delivery. Mol BioSystems. 2012; 8: 753759. DOI: 10.1039/c1mb05337j.

8. Liang P, Liu C, Zhuo R, Cheng S. Self-assembled inorganic/organic hybrid nanoparticles with multi-functionalized surfaces for active targeting drug delivery. J Mat Chem B. 2013; 1: 4243-4250. DOI: 10.1039/C3TB20455C.

9. Trushina DB, Akasov RA, Khovankina AV, Borodina TN, Bukreeva TV, Markvicheva EA. Doxorubicin-loaded biodegradable capsules: Temperature induced shrinking and study of cytotoxicity in vitro. J Mol Liquids. 2019; 284: 15215-224. DOI: 10.1016/j.molliq.2019.03.152.

10. Binevski PV, Balabushevich NG, Uvarova VI, Vikulina AS, Volodkin D. Bio-friendly encapsulation of superoxide dismutase into vaterite CaCO3 crystals. Enzyme activity, release mechanism, and perspectives for ophthalmology. Colloids and Surfaces B: Biointerfaces. 2019; 181: 437-449. DOI: 10.1016/j.colsurfb.2019.05.077.

11. Parakhonskiy B, Tessarolo F, Haase A, Antolini R. Dependence of sub-micron vaterite container release properties on pH and ionic strength of the surrounding solution. Adv Sci Technology. 2013; 86: 81-85. DOI: 10.4028/www.scientif-ic. net/AST.86.81.

12. Ogino T, Suzuki T, Sawada K. The formation and transformation mechanism of calcium carbonate in water. Geo-chim Cosmochim Acta. 1987; 51(10): 2757-2767.

13. Sergeeva A, Sergeev R, Lengert E, Zakharevich A, Par-akhonskiy B, Gorin D, Sergeev S, Volodkin D. Composite magnetite and protein containing CaCO3 crystals. External manipulation and vaterites-calcite recrystallization-mediat-ed release performance. ACS Appl Mater Interfaces. 2015; 7:21315-21325. DOI: 10.1021/acsami. 5b05848.

14. Sudareva NN, Saprykina NN, Popova EV, Vilesov AD. Porous calcium carbonate cores as templates for preparation of peroral proteins delivery systems. The influence of composition of simulated gastrointestinal fluids on the structure and morphology of carbonate cores. Chapter 4. In: "Calcium Carbonate: Occurrence, Characterization and Applications". (Ed. A.Cohen), Nova Science Publishers, Inc (NOVA). 2015, pp.73-95.

15. Sudareva N, Suvorova O, Saprykina N, Smirnova N, Bel'tiukov P, Petunov S, Radilov A, Vilesov A. Two-level delivery systems based on CaCO3 cores for oral administration of therapeutic peptides. J Microencapsulation. 2018; 35: 619634. DOI:10.1080/02652048.2018.155924.

16. Sudareva N, Popova H, Saprykina N, Bronnikov S. Structural optimization of calcium carbonate cores as templates for protein encapsulation. J Microencapsulation. 2014; 3(14): 333-343. DOI: 10.3109/02652048.2013.858788.

17. Parakhonskiy BV, Haase A, Antolini R. Sub-micrometer vaterites containers: synthesis, substance loading, and release. Angew Chem Int. Edition. 2012; 51: 1195-1197.

18. Zou Z, Bertinetti L, Politi Y, Fratzl P, Habraken WJ. Control of polymorph selection in amorphous calcium carbonate crystallization by poly(aspartic acid): two different mechanisms. Small. 2017; 13:1603100. DOI: 10.1002/ smll.201603100.

19. Nagaraja AT, Pradhan S, McShane MJ. Poly(vinylsulfon-ic acid) assisted synthesis of aqueous solution stable vater-ite calcium carbonate nanoparticles. J Colloid Interface Sci. 2014; 418: 366-372.

20. Zhao D, Zhuo R, Cheng S. Alginate modified nanostruc-tured calcium carbonate with enhanced delivery efficiency for gene and drug delivery. Mol Biosystems. 2012; 8:753-759.

21. Gusliakova O, Atochina-Vasserman EN, Sindeeva O, Sindeev S, Pinyaev S, Pyataev N, Revin V, Sukhorukov GB, Gorin D, Gow AJ. Use of submicron vaterite particles serves as an effective delivery vehicle to the respiratory portion of the lung. Front Pharmacol: Exp Pharmacol and Drug Discovery. 2018; 9: Article 559. DOI: 10.3389/fphar.2018.00559.

22. Borodina, TN, Trushina DB, Marchenko IV, Bukreeva TV. Calcium carbonate-based mucoadhesive microcontainers for intranasal delivery of drugs bypassing the blood-brain barrier. BioNanoScience. 2016; 6: 261-268. DOI: 10.1007/ s12668-016-0212-2.

23. Genina EA, Svenskaya YI, Yanina IY, Dolotov LE, Navo-lokin NA, Bashkatov AN, Terentyuk GS, Bucharskaya AB, Maslyakova GN, Gorin DA, Tuchin VV, Sukhorukov GB. In vivo optical monitoring of transcutaneous delivery of calcium carbonate microcontainers. Biomed Opt Express. 2016; 7: 2082-2087. DOI: 10.1364/BOE.7.002082.

24. Ohgushi H, Okumura M, Yoshikawa T, Inboue K, Senpu-ku N, Tamai S, Shors EC. Bone formation process in porous calcium carbonate and hydroxyapatite. J Biomed Mater Res. 1992; 26 (7): 885-895. DOI: 10.1002/jbm.820260705.

25. Trushina DB, Bukreeva TV, Kovalchuk MV, Antipina MN. CaCO3 vaterite microparticles for biomedical and personal care applications. Materials Sci Engineering C. 2014; 45: 644-658. DOI: 10.1016/j.msec.2014.04.050.

26. Parakhonskiy B, Zyuzin MV, Yashchenok A, Carre-gal-Romero S, Rejman J, Möhwald H. The influence of the size and aspect ratio of anisotropic, porous CaCO3 particles on their uptake by cells. J Nanobiotechnol. 2015; 13: Article 53. doi: 10.1186/s12951-015-0111-7.

Ватериты CaCO3 как компоненты системы направленной доставки лекарственных препаратов

iНе можете найти то, что вам нужно? Попробуйте сервис подбора литературы.

Наталья Н. Сударева 1'2, Павел В. Попрядухин 1'2, Наталья Н. Сапрыкина Ольга М. Суворова Галина Ю. Юкина 2, Олег В. Галибин 2, Александр Д. Вилесов 1,2

1 Институт высокомолекулярных соединений РАН, Санкт-Петербург, Россия

2 Первый Санкт-Петербургский государственный медицинский университет им. акад. И. П. Павлова, Санкт-Петербург, Россия

Резюме

Успехи в лечении большинства онкологических заболеваний связаны с применением действенных и в различной мере токсичных противоопухолевых препаратов. Во многих случаях химиотерапия требует максимальной локализации препарата в зоне опухоли. Поэтому наиболее эффективным методом является введение медицинских препаратов (МП) непосредственно в опухоль или применение тар-гетных систем их доставки. Второй из этих методов дает возможность снизить общую токсичность МП и достичь пролонгированного терапевтического эффекта из-за равномерного, контролируемого по времени выхода МП в опухолевую ткань.

В данной работе мы исследовали поведение пористых сферических частиц ватерита СаСО3 (компонентов системы доставки противоопухолевых препаратов) в различных средах (плазма крови человека,

мышечная ткань крыс). Было показано, что исследуемый носитель МП подвергается морфологической трансформции и со временем разрушается. В плазме крови, благодаря ионному обмену, ватериты превращаются в постепенно распадающиеся иглоподобные структуры, что показано с помощью сканирующей электронной микроскопии и энергорассеивающей спектроскопии. Сходные процессы наблюдались в мышечной ткани: в течение 3 дней сферические частицы превращались в иглоподобные структуры и затем подвергались полной биологической резорбции.

Ключевые слова

Противоопухолевые препараты, системы доставки, СаСО3, ватериты, плазма крови, внутримышечное введение, биорезорбция.

i Надоели баннеры? Вы всегда можете отключить рекламу.