Научная статья на тему 'Cancer diagnostics, imaging and treatment by nanoscale structures targeting'

Cancer diagnostics, imaging and treatment by nanoscale structures targeting Текст научной статьи по специальности «Биотехнологии в медицине»

CC BY
81
35
i Надоели баннеры? Вы всегда можете отключить рекламу.
Журнал
Biotechnologia Acta
CAS
Ключевые слова
CANCER DIAGNOSTIC / IMAGING AND TREATMENT / NANOPARTICLES / ДіАГНОСТИКА РАКУ / ВіЗУА ЛіЗА ЦіЯ ТА ЛіКУВАННЯ / НАНОЧАСТИНКИ / ДИАГНОСТИКА РАКА / ВИЗУАЛИЗАЦИЯ И ЛЕЧЕНИЕ / НАНОЧАСТИЦЫ

Аннотация научной статьи по биотехнологиям в медицине, автор научной работы — Özcan Öznur Özge, Karahan Mesut

Recent research focused on finding new strategies in cancer therapy that did not have significant side effects and was more effective than traditional modules including the surgical intervention, radiation and chemotherapeutics. In this regard the nanoscale structures provide useful approaches for cancer treatment. So, the nanoparticle systems improve the efficiency of therapeutic drugs reducing their side effects. Although many studies reported the development of novel cancer cell therapies for future, the clinical success is lacking understand the effects of nanoparticle type, size and dose with their usage areas. Thus, this review was aimed to illustrate the usage of nanoparticles in cancer diagnostic, imaging and treatment.

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.

ДИАГНОСТИКА, ВИЗУАЛИЗАЦИЯ И ЛЕЧЕНИЕ РАКА С ИСПОЛЬЗОВАНИЕМ НАНОРАЗМЕРНЫХ СТРУКТУР

Современные исследования направлены на поиск новых стратегий лечения рака, не имеющих значительных побочных эффектов и более эффективных по сравнению с традиционными методами, включая хирургическое вмешательство, лучевую терапию и химиотерапию. Применение наноразмерных структур дает возможность использовать полезные подходы для лечения рака.Системы наночастиц повышают эффективность терапевтических препаратов, снижая их побочные эффекты. Хотя во многих исследованиях сообщается о разработке новых методов лечения раковых клеток в будущем, клинический успех не позволяет понять влияние типа, размера и дозы наночастиц на области их применения. Таким образом, в настоящем обзоре проиллюстрирована возможность использования наночастиц в диагностике, визуализации и лечении рака.

Текст научной работы на тему «Cancer diagnostics, imaging and treatment by nanoscale structures targeting»

UDС 614.253 https://doi.org/10.15407/biotech12.06.012

CANCER DIAGNOSTICS, IMAGING AND TREATMENT BY NANOSCALE STRUCTURES

TARGETING

OZNUR OZGE OZCAN, MESUT KARAHAN Uskudar University, Istanbul, Turkey E-mail: mesut.karahan@uskudar.edu.tr

Received 10.09.2019 Revised 04.11.2019 Accepted 20.12.2019

Recent research focused on finding new strategies in cancer therapy that did not have significant side effects and was more effective than traditional modules including the surgical intervention, radiation and chemotherapeutics. In this regard the nanoscale structures provide useful approaches for cancer treatment. So, the nanoparticle systems improve the efficiency of therapeutic drugs reducing their side effects. Although many studies reported the development of novel cancer cell therapies for future, the clinical success is lacking understand the effects of nanoparticle type, size and dose with their usage areas. Thus, this review was aimed to illustrate the usage of nanoparticles in cancer diagnostic, imaging and treatment.

Key words: cancer diagnostic, imaging and treatment, nanoparticles.

Cancer continues to be one of the world's most devastating diseases with more than 10 million new cases each year [1]. The number of people diagnosed with malignancy is expected to rise to 22 million annually in the next 2 decades.

The information about various types of nanoparticles (NPs) used in cancer treatment, diagnosis and imaging was given under the title of various types of NPs used in the field of cancer treatment. After that, this review was focused on the active and passive targeting importance for the cancer apoptosis. In addition, the impact of Contemporary Advancements in active targeting nanoformulations were explained. In the last part, development and importance of different types of NPs in cancer diagnosis and imaging was discussed. Significance of this review is presentation in the field of cancer to date nanoparticles; a review with a holistic approach to their use in vaccines, drugs, diagnosis and imaging will be of great importance. Nanoparticles are very diverse and their efficacy and advantages vary according to the fields of medicine, vaccine and imaging. Which type of nanoparticles that we

are going to choose for our future studies could be seen in the reviews of our previous studies. Therefore, we aimed to present a work that we think it can be as useful as possible from the active, passive targeting of cancer to the varieties used in medicine and vaccines.

There is lack of understanding concerning the effects of the nanoparticle types, working in the future without confirming its reported function. This review will be useful for better comprehension of molecular basis of nanoparticles and the advent of new diagnostic technologies can help to improve the treatment of various cancers.

Various types of nanoparticles used in the field of cancer treatment

Treatment modalities, including immu-notherapy, photo thermal, photodynamic, gene and hormone therapy display promising cancer eradicating potential in preclinical studies. However, surgery, radiation and chemotherapy continue to be the first treatment option for cancers and the next major strategy for cancer treatment is highly non-specific in targeting the drugs to the cancer cells causing undesirable side-effects to the healthy tissues

[2-4]. Unfortunately, there are no alternative for cancer treatments other than surgical removal of the cancer site, radiotherapy and chemotherapy. In addition, chemotherapy and radiotherapy have thousands of side effects that kill both cancer cells and healthy cells. Therefore, we see that cellular therapies, which we believe will be more effective for cancer treatment, have started to be developed. In developing cellular therapies, nanocarriers that can be produced up to cellular dimensions have been of great importance. There are possible risks in these systems. For instance, NPs can be phagocytosed as an enemy in the body by being exposed to attacks of immune cells and may cause failure of such cancer treatment intervention. In addition, NPs can produce molecular responses such as deleterious, allergic and toxic effects in cancer cells, thereby causing greater sensitivity to cancer progression [5]. For this reason, choosing the most accurate NPs for the cancer disease will prevent patients from complications and can provide more effective therapy. During the last decades, an abundance of NPs have been developed and a real hype has been created around their potential application as diagnostic and therapeutic agents. For example, although iron oxide NPs have been suggested as potential diagnostic agents, they have not been fully preferred for clinical purposes. Therefore, pre-clinical studies on many experimental animals are ongoing [6]. This is primarily due to the ability of NPs to be biologically suitable for the body. There are some problems related to biological degradation of NPs, expulsion from the body and the possibility of toxic effects. Further studies are needed to eliminate these drawbacks of Iron oxide NPs.

Molecular imaging, when used in conjunction with said nanosystems, may make cancer diagnosis and treatment more effective. Multidisciplinary studies can be combined to provide more effective diagnosis and treatment, and these disciplines can often be made possible by collaborating with researchers in different fields such as cell biology, biochemistry, engineering, health and medicine. Recent advances in NPs technology have enabled the fabrication of NPs classes with unique sizes, shapes and materials, which in turn has facilitated major advancements in the field of nanomedicine. The promising proposition of multifunctional NPs for cancer diagnostics and therapeutics has inspired the development of the approach for improved cancer therapy. The nature of NPs is closely related with the various materials that used

for their synthesis, such as metals (gold and silver), ceramics (hydroxyapatite), lipids (cholesterol and non-toxic phospholipids) and polymers [alginate, chitosan, poly(ethylene glycol (PEG)] [7].

The combination of anti-cancer drugs and different types of agents with NPs in the treatment of cancer has many advantages:

1. Enhancing the stability of hydrophobic drugs, making them suitable for application. In other words, it increases the water solubility of drugs. Especially in imaging and drug therapies developed with these nanoscales, only cancer cells can be targeted and adverse effects such as systemic chemotherapy, hair loss, immunosuppression, muscle weakness, etc. can be completely eliminated.

2. Reducing toxicity by using biocompatible nanomaterials. In both conditions it results in a therapeutic index increase, the limit between doses causing therapeutic efficacy (e.g. cancer cell apoptosis) exhibits high differential uptake efficiency in the target cells over normal cells.

3. Developing pharmacokinetics and bio distribution which increase drug efficiency on the tumor and enhance absorption of the drugs into a selected tissue (for example, solid tumor) [8].

4. One of the most important advantages of the NPs is the fact that they are able to treat cancer which is caused by cell organelle disorder. For example, effective targeting of mitochondria and nucleus has emerged as an alternative strategy in cancer chemotherapy agent which is Dual drug conjugated NP [9].

Nevertheless, before the plethora of nanodevices currently under investigation become proper for clinical usage, they have to pass the rigorous tests set forth by regulatory agencies such as Food and Drug Administration (FDA) and European Medicines Agency (EMEA) [10-12]. To date, at least 12 polymer-drug conjugates have entered Phase I and II clinical trials and we need to find more treatment model for cancer cell targeted therapy. Various biocompatible (NPs)-based drug-delivery systems such as liposomes, dendrimers, micelles, silica, quantum dots, and magnetic, gold, and carbon nanotubes have already been reported for targeted cancer treatment.

Silver Nanoparticles (AgNPs)

Therapeutic applications of silver nanoparticles (AgNPs) agents in the diagnosis and probing of many cancer diseases are noteworthy [13]. AgNPs cause apoptosis in

cancer cells as they produce reactive oxygen species (ROS) that cause oxidative stress and DNA damage that cause mitochondrial damage within the cell. Cellular penetration of AgNPs usually occurs through endocytosis [14]. In another study, it was found that AgNPs for cell morphology may affect the function of other factors. These effects include the ability to adsorb cytosolic proteins and regulate gene expression and proinflammatory cytokines, for example, microarray analysis showed that human lung epithelial cell line A549 affects cellular transcriptome analysis upon exposure to AGNPs. According to the results of the microarray study, it was found that AgNPs affect the regulation of more than 1000 genes

[15]. AgNPs can induce autophagy by allowing the accumulation of autophagolysis in human ovarian cancer cells and autophagy can have a dual cell bodies. The use of autophagy inhibitors or autophagy protein 5 (ATG5) with small-mix RNAs (siRNA) in combination with AgNPs causes the death of cells in cancer cells

[16]. As a result, AgNPs can induce cell death including ROS generation, leakage of lactate dehydrogenase, increasing of apoptosis and autophagy genes, cause endoplasmic reticulum stress and mitochondrial damage, activation of caspases pathways and DNA damage so that AgNPs can be used as nanoparticle that is significant to deliver drugs for cancer treatment. In addition, AgNPs has significant importance to modulate ABC transporter activity for chemotherapy in multidrug resistant cancer [17].

Gold nanoparticles

Gold nanoparticles (AuNPs) are ideal for drug targeting and also for imaging-based detection of cancer diseases at an early stage. AuNPs were first produced in 1857 by Faraday and exhibit favorable physical properties and tailored surface functionalization, providing a potential for developing cancer theranostics and they are solid balls of gold and are made by the reduction of chloroauric acid, and their diameter varies from 5 to 100 nm. They are biocompatible and less toxicity and display the relatively low rate of clearance from circulation. AuNPs was demonstrated [18] where AuNPs were conjugated with an antibody against the epidermal growth factor receptor (EGFR, it is known to overexpress on many cancers). PEGylation of AuNPs effectively downregulate this uptake by macrophages and monocytes [19]. AuNPs conjugated with carbohydrates and proteins have been utilized in novel approaches

toward the development of vaccines such as Glyco-conjugated AuNPs (1-5 nm) capped with carbohydrate-based antigens that are present in cancer cells [20, 21]. Targeting mitochondria with conjugated Au- cationic NPs maltotriose-modified poly(propylene imine) (PPI) dendrimers effects on apoptosis induction in the human breast cancer cell line [22]. Doxorubicin (DOX) loaded oligonucleotides attached to (AuNPs) as a drug delivery system is useful for cancer chemotherapy [23]. Micro RNA (miR)-375 loaded AuNP exhibits high cellular uptake and preserves miR-375's activities to suppress cellular proliferation, migration/invasion, and colony formation, and to induce apoptosis in hepatocellular carcinoma cells [24].

Polymeric particles

Synthetic polymers are polylactic acid (PLA), poly(lactic-co-glycolic acid (PLGA), polycaprolactone (PCL), PEG, and poly(vinyl alcohol (PVA) are composed of commonly used natural polymers. Chemotherapeutic drugs (Trastuzumab, Pentuzumab, Paclitaxel, DOX, 5-Fluorouracil and Dexamethasone, etc.) become more effective when they are encapsulated with polymeric NPs.

NPs synthesized from PLGA, a synthetic polymer, are widely studied for anticancer drugs [26, 27]. PLGA has several advantages over cell-targeted therapies compared to other delivery systems:

1) It has been approved by the FDA for drug delivery in humans [28, 29].

2) Biodegradable.

3) Has sustained release activity, ranging from days to weeks under physiological conditions.

4) Provides long-term stability of charged bioactive molecules.

5) Shows ability to capture hydrophobic and hydrophilic drugs.

6) Has comprehensive functionalization options.

PEG is a water-soluble, biocompatible polymer commonly used for coating a wide variety of drugs to improve encapsulation efficiency. Especially in Human Epidermal Receptor 2 (HER2)-related breast cancer, mir-21 is effective in tumor immunity of Antisense Oligonucleotides (ASO) [30]. It has been demonstrated in the animal model that the antitumor effect is quite high even though MiR-21 was used previously only with targeting ligand by PLGA-PEG encapsulation of ASO [26, 31-33]. A biodegradable poly (D, L-lactide-co-glycolide) -block-poly (ethylene glycol) (PLGA-b-PEG-COOH) copolymer will be

synthesized. The most important factor in PEG expression is the prevention of immune system agents [34]. The strong buffering capacity of cationic polymers could effectively help themselves to escape from endo/lysosome as a result of "proton sponge" effect. For instance, 25 kDa polyethylenimine is well known of its excellent transfection activity in vitro largely due to its strong buffering capacity and these NPs is very useful for cancer immunotherapy for vaccinations [35-38].

Superparamagnetic iron oxide (SPIO) nanoparticle

Chemotherapeutic agents have been associated with SPIO-based nanocarriers through different strategies (e.g., conjugation via cleavable linker and %-% stacking with polymer layers) for delivery to tumors. Dual paclitaxel (PTX)/superparamagnetic iron oxide (SPIO)-loaded PLGA-based NPs have a potential role in tumor growth [39] in passive targeting.

"iRGD" peptide affects the uptake of iron oxide during labeling of pancl cells for this reason an appropriate "iRGD" peptide concentration enhances the uptake of intracellular iron tumor cell proliferation in active targeting [40]. Trastuzumab is conjugated to SPIO NPs which labor as

magnetic resonance imaging (MRI) contrast agents to detect HER2-positive tumors [41, 42].

Carbon nanotubes (CNTs)

Carbon NanoTubes (CNTs) are carbon allotropes with a cylindrical nanostructure which have gained intensive interest during the past 20 years because of their unique mechanical properties in addition to very interesting values in electrical and thermal conductivity and also the possibility of their surface to functionalize with a wide group or biochemical species paving the way for numerous therapeutic and drug delivery applications [43, 44]. They are able to penetrate easily through the cellular membrane and have low immunogenicity with significant uptake of delivered small interfering RNA (siRNA) and a working gene silencing effect in the tumor tissue [45]. Many of them have high general toxicity and additional drawbacks, like limited solubility and a poor non-selective biodistribution [46].

Liposomes

Liposomes are broadly used as drug delivery systems and several liposomal nanomedicines have been approved for clinical applications. Liposome-based combination chemotherapy contributes a novel avenue in

fttnidttyp* CcnipMiti«ftr$tftittur* PrOptRÎH AppEutien*

9 poyrw ».g., PLCîA. çficmot, diisua DMA; itvansenitt, atpovn» n, hydef M Sem* bodtgrtdib'* Drug Mntry. pavjt Wl&M. ur>№1*d 'e'tdl* iliçfm*It

DtnAnw f'AMjVU. ttt low p&ydaen ty, c*go. bcccmptujt Drug dtüvtry

1 IpOWi«, m«*« Cfnony hydfw<iQt"C t-njc, 0*xa~\prtto. typcttr 50-500mi OuadtWy

Ol Outrtbm data CöSa. CiAvS*. CdTt. *k Bfted lnûumrt. rw MM «n, $-100 im

fold Spfwn. roijt. « ihils Bocornpttitpi (y. 5-KHJnm Hvpirt-iht i Ihwpy. dnjgiMwtrY

Sphrw. th»l), rrnwwi bflwtiwtoiîy CeriiH jjirij. dmg 4tt«iry (tflcapvjUtonJ

# Mjgwtc troncsod* « cottt'ï-tuféd. splw«. atxin m téct Supocivinusnfjc. <tti»Mgn>tic [wrul iiminru le m™ f-tOnml. fmwiwgnttc_ Conflit I!/SI. hyp*rih*?i?vi

\i \ Cvbotbtud Cttbon MWiubtt. budcyfak. 3) grifiwe B*i«r«wtiU* dilwtfy

Fig. 1. NPs types and their application areas for cancer imaging and treatment [25]

drug delivery research and has increasingly become a significant approach in clinical cancer treatment. Liposomes are grouped into two types:

1) unilamellar vesicles and

2) multilamellar vesicles.

For the treatment of multidrug resistance (MDR)/cancer immunotherapy, mixtures of siRNA/plasmid DNA and hydrophobic drug can be used with Liposomes [47].

Paclitaxel and Rapamycin (with antitumor and immunosuppressant properties and an inhibitor of mTOR protein kinase) are encapsulated with PEG-Liposome in breast cancer — Liposomes released in slow and sustained fashion | Cell line cytotoxicity | In vivo therapeutic effects. The system controlled the tumor growth [48].

Folate receptor, a membrane-associated folate binding protein, is overexpressed in over 90% of ovarian cancer and other epithelial types of cancer [49, 50]. Xu et al. developed a FR targeted co-delivery formulation by folate-DOX/Bmi1 siRNA liposome (FA-DOX/siRNA-L), demonstrated a great tumor targeting effect and prevented tumor growth in vitro and in vivo experiments [51, 52].

A thermosensitive magnetic liposomal delivery system is effective co-delivery of gene silencing short hairpin RNA (shRNA) vector and antitumor drug (DOX) into gastric cancer [53]. Liposomal system with an antimicrobial peptide and co-delivery of antagomir-10b could trigger cell death in the meantime besides hindering of T cells migration [54].

Polimeric Micelles

Polymeric micelles used due to their ability to load therapeutics, deliver the cargo to the site of action, improve the pharmacokinetic of the loaded drug and reduce off-target cytotoxicity. They are also developed with improved drug loading capabilities by modulating hydrophobicity and hydrophilicity of the micelle forming block co-polymers and also cancer targeted by surface modifying with tumor-homing ligands. Their classroom contains in Polymeric micelles of therapeutic applications in cancer treatment.

Pluronics®

PEG-PLA

PEG-PCL

PEG-Lipid

PEG-PLGA

CoDirnlloD*! liposome

A

D

PEGylalrd lipmomr

^»K.Tkit™- CtiioJ

fftci

B

Mullltuotliooal Ijpijiouii L(. thtrumlir tposoint

Sail

Ligaad targeted hpoiomr

Fig. 2. Illustration of the versatile functions of liposomes:

A — Conventional liposomes are composed of two layers of phospholipids and when administered intravenously and remain in the circulatory system for a short time. Also they are widely used for the maintenance of hydrophilic drugs that did not pass through the cell membrane; B — When protected with PEG, they have the ability to be protected from immune cell attacks and stay longer

in circulation;

C — When liposomes are used for active targeting with small molecules such as aptamer, peptide, ligand and

protein, they show very high effect on cancer cells; D — Especially in the diagnosis and treatment of solid tumors they have teranostic effects. They are also often

preferred for imaging (such as MRI) [55]

6. PEG-poly(amino acids)

7. Stimuli-sensitive polymeric micelles

8. Endogenous stimuli-sensitive polymeric micelles

9. pH-sensitive polymeric micelles

10. Reduction sensitive polymeric micelles

11. Thermo-sensitive polymeric micelles

12. Exogenous stimuli-sensitive polymeric micelles

13. Light-sensitive polymeric micelles

14. Magnetic field-sensitive polymeric micelles

15. Ultra-sound sensitive polymeric micelles

16. Margination of micro/nanoparticles: Requirement for optimum drug delivery.

Other hydrophilic block forming polymers include chitosan, poly(N-vinyl pyrrolidone) (PVP), and poly(Nisopropylacrylamide) (pNIPAAm). There are various polymer blocks used to form micellar core, including the class of polyethers such as poly(propylene oxide) (PPO), various polyesters such as PLA, PCL, PLGA, poly(P-aminoesters), polyamino acids such as poly(L-histidine) (pHis), poly(L-aspartic acid) (pAsp) and lipids such as dio leoyl(phosphatidylethanolamine) (DOPE), distearoyl (phosphatidylethanolamine) (DSPE). The assembly of block co-polymers, in which PPO attached to PEG as A-B-A triblock co-polymers (PEO-PPO-PEO) is known as Pluronics [56].

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

Protein Nanoparticles

Protein NPs are also used including water-soluble proteins (e.g., bovine and human serum albumin) and insoluble proteins (e.g., zein and gliadin). So far, most of proteins NPs of article are focused on the preparation and

characterization of nanoparticles derived from gelatin, albumin, gliadin, legumin, Methoxy-PEG-polylactide, PEG-asparaginase and two milk proteins that have been investigated for drug delivery applications are Beta-lactoglobulin (BLG) and casein [57, 58]. To promote drug targeting ability, protein nanoparticles have been chemically modified to incorporate targeting ligands that recognize specific cells and tissues. Such modification allows targeting of albumin nanoparticles to breast cancer cells, which overexpress HER2 [59]. Gliadin NPs used as a bioactive delivery system for oral vaccines administration to aid the sustained release delivery of anticancer drugs as well as colon cancer-targeted cyclophosphamide drug therapy and effective for apoptosis of breast cancer cells [60]. Cisplatin-loaded casein is a milk protein nanoparticles demonstrated their ability to penetrate cell membranes, target tumors, and inhibit tumor growth in hepatic tumor [61].

Cancer Treatment with Active and Passive Targeting

Active Targeting Strategies

NPs are used for active targeting [71] to cancer cells including antibody and antigen, peptide, protein, aptamer and ligand fragment based targeting in figure 3 [64]. HER2 is overexpressed in approximately 25-30% of invasive breast cancer but is less expressed by normal adult tissues. Targeted treatment with humanized Trastuzumab (Monoclonal Antibody) targeting the HER2 receptor has become the mainstay of HER2 positive breast cancer. The significant effect of Trastuzumab-conjugated nanoparticles to specifically target HER2 positive cancer cells has been

Fig. 3. Illustration of active targeting components in cancer cells [62]

proved in vitro using different cell lines and in vivo [62]. The ZHER2 antibody protein molecule consisting of 58 amino acids and 3 domains is designed as a high affinity linker to HER2 receptors. ZHER2 binds to the HER2 receptor, a domain point different from the point at which the therapeutic antibodies of Trastuzumab and Pertuzumab bind [63]. For this reason ZHER2 protein is highly used for diagnosis (imaging) and treatment of cancer diseases [64, 65]. Cetuximab has been targeted specifically and efficiently AuNPs in Epidermal Growth Factor Receptor (EGFR)-positive pancreatic and colorectal carcinoma cell lines [66].

Passive targeting facilitates the efficient localization of NPs in tumor interstitium but cannot further promote their uptake by cancer cells [71]. Uptake can be achieved by actively targeting NPs to receptors or other surface membrane proteins overexpressed on target cells. The addition of targeting ligands allows the delivery of drug-encapsulated NPs to uniquely identifiable cells or even subcellular sites, thereby reducing the unwanted systemic exposure of cytotoxic drug. Specific interactions between the ligands on the surface of nanocarriers and receptors expressed on the tumor cells may facilitate NPs internalization by triggering receptor-mediated endocytosis. Furthermore, active targeting of nanocarriers with small molecule therapeutic cargo has shown the potential to suppress multidrug resistance (MDR) via bypassing of P-glycoprotein-mediated drug efflux [67, 68]. As a result, although passive targeting facilitates the effective localization of NPs in the tumor interstitium, it cannot over-stimulate cellular uptake by cancer cells, such as active targeting.

NPs are functionalized with different biological molecules, peptides, antibody, and protein ligands for targeted drug delivery and also contain non-coding RNA, viral [69] and bacteria's DNA [38] or RNA for cancer immunization and cell death progress. Natural plant-based drugs that can be used in the field of pharmacology have also been found to be more effective when used with NPs such as Prosopis Cineraria. It is a leaf located in India, which can be used as NP-plant-based drug system for cancer treatment [70]. Ligands for active targeting in drug delivery approach on its great affinity to somatostatin receptors (SSTRs), which is overexpressed in several cancer cells such as core-shell type liposome co-encapsulating VEGF-targeted siRNA (siVEGF) is very effective drug system for VEGF based

cancer types. SPIO NPs are accumulate in cancer through passive targeting by the EPR effect and with active targeting to help of targeting by ligands.

Passive Targetting Strategies

Although NPs refer to accumulate in the tumor cells due to the enhanced permeation and retention (EPR) effect, passive tumor targeting is dependent on the tumor vascularization and angiogenesis, and therefore lacks specificity and consistency. Both the tumor model type and conditions can seriously affect the passive targeting effectiveness [72-74]. General features of tumors include leaky blood vessels and

Fig. 4. An example for Aptamer modeling to target cancer cells (generally receptors, ligands, etc.) [25]

poor lymphatic drainage. Free drugs may be used nonspecically as a nanocarrier and can extravagate into the tumor tissues via the leaky vessels by the EPR effect. Sometimes, targeting cells within a tumor is not always feasible because some drugs cannot be used efficiently and the random nature of the approach makes it difficult to control the process which is lack of control may induce MDR.

Nanoparticle Imaging System

Imaging techniques used mostly in preclinical studies and clinical practice such as MRI, computed tomography (CT), ultrasound (US), optical imaging (OI), photoacoustic imaging (PAI), positron emission tomography (PET), and single-photon emission Computed tomography (SPECT).

Targeted NPs imaging agents provide a new technology for cancer imaging, which goes beyond anatomical characterization. It enables early detection of cancer as well as treatment monitoring at the molecular and cellular level. With the development of nanotechnology, magnetic NPs have been used in the MRI, adhering to target cells and drug release system [75, 76]. Tumor diagnosis and treatment can be obtained by the same NPs formulation and the disease can be monitored and fought by the synergistic effect of more than one therapy. The preparation and application of single multifunctional nano radiotracers based on iron oxides and enabling PET/MRI dual imaging can used for treatment and diagnosis for cancer [77]. New designed and realized a bimodal CT for SPECT and MRI with Superparamagnetic Iron Oxide NPs (SPION) privileged the magnetic properties to the CNT, while 99mTc granted the radioactive property.

Gold Nanoparticle Imaging

AuNPs are now used widely in bioimaging and phototherapy due to their tunable properties and highly sensitive optical and electronic properties of the surface plasmon resonance (SPR) [78]. AuNPs act as an active imaging probe for cancer detection facilitating whole-body scans. AuNPs can be easily functionalized with additional imaging agents by improving the AuNP-based imaging systems. That may allow the observation of tissues not only on its basic anatomic configuration but also on the molecular level for cancer diseases. For example, Au atoms using a one-step procedure for SPECT/CT imaging in an orthotropic mouse xenograft of

triple-negative breast cancer (TNBC) and also PEGylation for favorable pharmacokinetics and d-Ala1-peptide T-amide (DAPTA) for targeting C-C chemokine receptor 5 (CCR5, a prognostic biomarker for breast cancer progression) [79, 80]. Multifunctional gold nanoprobe is designed for simultaneous miRNA-21 responsive fluorescence imaging and therapeutic monitoring of cancer. miRNAs provides a simple but powerful protocol with great potential in cancer imaging, therapy, and therapeutic monitoring [81].

Superparamagnetic iron oxide (SPIO) Imaging

Superparamagnetic iron oxide (SPIO) NPs were studied for the development of contrast agents in MRI for cancer diagnosis. First-generation of SPIO NPs had diagnostic capabilities only, whereas a new model of SPIO NPs has multifunctional characteristics for combined therapeutic and diagnostic applications for cancer. The magnetite (Fe3O4) and maghemite (Fe2O3) cores of SPIO NPs can be readily detected with MRI, thereby enabling real-time in vivo drug tracking. To provide colloidal stability of the magnetic core and better biocompatibility, SPIO NPs have been stabilized with polysaccharides (e.g., dextran and chitosan), PEG, polypyrrole (PPy), PLA, PLGA and their copolymers. Compared with other coating materials such as silica, polymer advantages with great biocompatibility and biodegradability to facilitate MRI-guided drug delivery, gene delivery, photo thermal therapy (PTT), photodynamic therapy 5 (PDT) or magnetic hyperthermia [82] growth through the EPR effect and available real-time in vivo drug tracking with MRI [83].

Discussion

Current cancer treatments include surgical intervention, radiation and chemotherapeutic agents. Side effects are an integral part of these treatment modules. So, recent studies focused on finding new strategies without any major side effects and were more effective instead of these modules. However, better comprehension of molecular basis of tumor and the advent of new diagnostic technologies (such as active and passive targeting models) and treatments can be decreased mortality rate in cancer patients. So the researchers continue to find strategies to improve the chances of survival and quality of cancerous patient's lives. NPs are used in the search for this new treatment method and can be preferred in imaging methods as well [84].

Nanoparticles and related biotechnologies provide needed augmented presentation for development of vaccine, treatments, diagnosis, imaging active and passive strategies for cancer and undoubtedly nanoparticle engineering. For this purpose an exciting ongoing and future studies and an increasing focus of clinical trials cancer treatments will remain.

Consequently, this review provides a brief manual for anyone in the field of nanotechnology for the diagnoses, treatment and vaccination of the cancer disease.

This work was supported by the Usktidar University.

REFERENCES

1. Stewart B. W., Kleihues P. World Cancer Report. World Health Organization Press. Avaible at https://publications.iarc.fr/Non-Series-Publications/World-Cancer-Reports/ World-Cancer-Report-2003 (accessed, Geneva, 2003).

2. Jemal A., Siegel R, Xu J., Ward E. Cancer statistics. Cancer J. Clin. 2010, V. 60, P. 277-300.

3. Peer D, Karp J. M, Hong S, Farokhzad O. C, Margalit R., Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007, V. 2, P. 751-60.

4. Kumari P., Ghosh B., Biswas S. Nanocarriers for cancer-targeted drug delivery. J. Drug Targeting. 2015, 24 (3), 179-191. https://doi/ org/10.1038/nnano.2007.387

5. Kononenko V., Narat M., Drobne D. Nanoparticle interaction with the immune system Arch. Industr. Hygiene Toxicol. 2015, 66 (2), 97-108. https://doi/org/10.1515/aiht-2015-66-2582

6. Baetke S. C., Lammers T., Kiessling F. Applications of nanoparticles for diagnosis and therapy of cancer. Brit. J. Radiol. 2015, 88 (1054), 20150207. https://doi/ org/10.1259/bjr.20150207

7. Yih T, Al-Fandi M. Engineered nanoparticles as precise drug delivery systems. J. Cel. Biochem. 2006, V. 97, P. 1184-1190. https:// doi/org/10.1002/jcb.20796

8. Jabir N. R., Tabrez S., Ashraf G. M., Shakil S., Damanhouri G. A., Kamal M. A. Nanotechnology-based approaches in anticancer research. Int. J. Nanomedicine. 2012, V. 7, P. 4391. https://doi/org/10.2147/ IJN.S33838

9. Mallick A., More P., Ghosh S., Chippalkatti R., Chopade B. A., Lahiri M., Basu S. Dual Drug Conjugated Nanoparticle for Simultaneous Targeting of Mitochondria and Nucleus

in Cancer Cells. ACS Applied Materials & Interfaces. 2015, 7 (14), 7584-7598. https:// doi/org/10.1021/am5090226

10. Stammati A. P., Silano V., Zucco F. Toxicology investigations with cell culture systems. Toxicology. 1981, V. 20, P. 91-153.

11. Borm P., Klaessig F. C., Landry T. D., Moudgil B., Pauluhn J. Research strategies for safety evaluation of nanomaterials, part V: role of dissolution in biological fate and effects of nanoscale particles. Toxicol. Sci. 2006, V. 90, P. 23-32. https://doi/org/10.1093/toxsci/ kfj084

12. Costa E. C., Gaspar V. M., Marques J. G, Coutinho P., Correia I. J. Evaluation of Nanoparticle Uptake in Co-culture Cancer Models. PLoS ONE. 2013, 8 (7), e70072. https://doi/org/10.1371/journal. pone.0070072

13. ZhangX.-F., Liu Z.-G., Shen W., Gurunathan S. Silver Nanoparticles: Synthesis, Characterization, Properties, Applications, and Therapeutic Approaches. Inter. J. Mol. Sci. 2016, 17 (9), 1534. https://doi/ org/10.3390/ijms17091534

14. AshaRani P. V., Mun G. L. K., Hande M. P., Valiyaveettil S. Cytotoxicity and genotoxicity of silver nanoparticles in human cells. ACS Nano. 2009, V. 3, P. 279-290. https://doi/ org/10.1021/nn800596w.

15. Foldbjerg R., Irving E. S., Hayashi Y., Sutherland D. S., Thorsen K., Autrup H., Beer C. Global gene expression profiling of human lung epithelial cells after exposure to nanosilver. Toxicol. Sci. 2012, V. 130, P. 145-157. https://doi.org/10.1093/ toxsci/kfs225

16. Lin J., Huang Z., Wu H., Zhou W., Jin P., Wei P., Zhang Y., Zheng F., Zhang J., Xu J. Inhibition of js_E_piautophagy enhances the anticancer activity of silver nanoparticles. Autophagy. 2014, V. 10, P. 2006-2020. https://www.ncbi.nlm.nih.gov/pmc/ articles/PMC6454023

17. Kovacs D., Szoke K., Igaz N., Spengler G., Molnar J., Toth T., Kiricsi M. Silver nanoparticles modulate ABC transporter activity and enhance chemotherapy in multidrug resistant cancer. Nanomed. Nanotechnol., Biol. Med. 2016, 12 (3), 601-610. https://doi. org/10.1016/j.envpol.2019.113880

18. Sokolov K., Follen M., Aaron J., Pavlova I. Real-time vital optical imaging of precancer using anti-epidermal growth factor receptor antibodies conjugated to gold nanoparticles. Cancer Res. 2003, 63 (9), 1999-2004. https://www.ncbi.nlm.nih.gov/pmc/ articles/PMC2773158

19. Kah J. C., Wong K. Y., Neoh K. G., Song J. H., Fu J. W., Mhaisalkar S. Critical parameters in the pegylation of gold nanoshells for

biomedical applications: An in vitro macrophage study. J. Drug Target. 2009, V. 17, P. 181-193. https://doi. org/10.1080/10611860802582442

20. Svarovsky S. A., Szekely Z, Barchi J. J. Synthesis of gold nanoparticles bearing the thomsen-friedenreich disaccharide: A new multivalent presentation of an important tumor antigen. Tetrahedron Asymmetry. 2005, V. 16, P. 587-598. https://doi. org/10.1016/j.tetasy.2004.12.003

21. Ojeda R, de Paz J. L, Barrientos A. G., Martin-Lomas M., Penades S. Preparation of multifunctional glyconanoparticles as a platform for potential carbohydrate-based anticancer vaccines. Carbohydrate Res. 2007, V. 342, P. 448-459. https://doi. org/10.1016/j.carres.2006.11.018

22. Mkandawire M. M., Lakatos M., Springer A., Clemens A., Appelhans D, Krause-Buchholz U., Mkandawire M. Induction of apoptosis in human cancer cells by targeting mitochondria with gold nanoparticles. Nanoscale. 2015, 7 (24), 10634-10640. https://doi. org/10.1039/C5NR01483B

23. Lee C. S., Kim H., Yu J., Yu S. H., Ban S., Oh S., Jeong D., Im J., M. J., Kim T. H. Doxorubicin-loaded oligonucleotide conjugated gold nanoparticles: A promising in vivo drug delivery system for colorectal cancer therapy. Europ. J. Med. Chem. 2017, V. 142, P. 416-423. https://doi. org/10.1016/j.ejmech.2017.08.063

24. Chang Y., Yan W., He X., Zhang L., Li C., Huang H., Nace G., Geller D. A., Lin J., Tsung A. miR-375 inhibits autophagy and reduces viability of hepatocellular carcinoma cells under hypoxic conditions. Gastroenterol. 2012, V. 143, P. 177-187 e8. https://doi. org/10.1053/j.gastro.2012.04.009

25. Dawidczyk C. M., Russell L. M., Searson P. C. Nanomedicines for cancer therapy: state-of-the-art and limitations to pre-clinical studies that hinder future developments. Frontiers in Chemistry. 2014, V. 2. https:// doi.org/10.3389/fchem.2014.00069

26. Devulapally R., Sekar N. M., Sekar T. V., Foygel K., Massoud T. F., Willmann J. K., Paulmurugan R. Polymer Nanoparticles Mediated Codelivery of AntimiR-10b and AntimiR-21 for Achieving Triple Negative Breast Cancer Therapy. ACS Nano. 2015, 9 (3), 2290-2302. https://doi.org/10.1021/ nn507465d

27. Fernandez-Fernandez A., Manchanda R., McGoron A. J. Theranostic Applications of Nanomaterials in Cancer: Drug Delivery, Image-Guided Therapy, and Multifunctional Platforms. Appl. Biochem. Biotechnol. 2011, V. 165, P. 1628-1651. https://doi. org/10.1007/s12010-011-9383-z

28. Lu J. M, Wang X., Marin-Muller C, Wang H, Lin P. H, Yao Q, Chen C. Current Advances in Research and Clinical Applications of PLGA-Based Nanotechnology. Expert Rev. Mol. Diagn. 2009, V. 9, P. 325-341. https:// doi.org/10.1586/erm.09.15

29. Mundargi R. C, Babu V. R., Rangaswamy V., Patel P., Aminabhavi T. M. Nano/Micro Technologies for Delivering Macromolecular Therapeutics Using Poly(D,LLactide-Co-Glycolide) and Its Derivatives. J. Control. Release. 2008, V. 125, P. 193-209.

30. Mattos-Arruda L., Giulia Bottai, Paolo G. Nuciforo, Luca Di Tommaso, Elisa Giovannetti, Vicente Peg, Agnese Losurdo, José Pérez-Garcia, Giovanna Masci, Fabio Corsi, Javier Cortés, Joan Seoane, George A. Calin, Libero Santarpia. MicroRNA-21 links epithelial-to-mesenchymal transition and inflammatory signals to confer resistance to neoadjuvant trastuzumab and chemotherapy in HER2-positive breast cancer patients. Oncotarget. 2015, V. 6, P. 37269-37280. https://doi.org/10.18632/oncotarget.5495

31. Malhotra M., Thillai Veerapazham Sekar, Jeyarama S. Ananta, Rammohan Devulapally, Rayhaneh Afjei, Husam A. Babikir, Ramasamy Paulmurugan, Tarik F. Massoud. Targeted nanoparticle delivery of therapeutic antisense microRNAs presensitizes glioblastoma cells to lower effective doses of temozolomide in vitro and in a mouse model. Oncotarget. 2018, V. 9, P. 21478-21494. https://doi.org/10.18632/ oncotarget.25135

32. Mohammadian F., Pilehvar-Soltanahmadi Y., Mofarrah M., Dastani-Habashi M., Zargha-mi N. Down regulation of miR-18a, miR-21 and miR-221 genes in gastric cancer cell line by chrysin-loaded PLGA-PEG nanoparticles. Artif. Cel., Nanomed., Biotechnol. 2016, 44 (8), 1972-1978. https://doi.org/10.3109 /21691401.2015.1129615

33. M ller V., Gade S., Steinbach B., Loibl S., von Minckwitz G., Untch M., Schwarzenbach H. Changes in serum levels of miR-21, miR-210, and miR-373 in HER2-positive breast cancer patients undergoing neoadjuvant therapy: a translational research project within the Geparquinto trial. Breast Cancer Research and Treatment. 2014, 147 (1), 61-68. https://doi.org/10.1007/s10549-014-3079-3

34. Harris J. M., Chess R. B. Effect of Pegylation on Pharmaceuticals. Nat. Rev. Drug Discov. 2003, V. 2, P. 214-221. https://doi. org/10.1038/nrd1033

35. Mintzer M. A. Simanek E. E. Non Viral Vectors for Gene Delivery. Chem. Rev. 2009, V. 109, P. 259-302. https://doi.org/ 10.4103/2277-9175.98152

36. Yin H., Kanasty R. L., Eltoukhy A. A., Vegas A. J., Dorkin J. R., Anderson D. G. Non

Viral Vectors for Gene-based Therapy Nat. Rev. Genet. 2014, V. 15, P. 541-555.

37. Pietersz G.A., Tang C. K., Apostolopoulos V. Mini Rev. Med. Chem. 2006, V. 6, P. 1285-1298.

38. Hu Q., Wu M., Fang C., Cheng C., Zhao M, Fang W., Tang G. Engineering Nanoparticle-Coated Bacteria as Oral DNA Vaccines for Cancer Immunotherapy. Nano Letters. 2015, 15 (4), 2732-2739. https://doi.org/10.1021/ acs.nanolett.5b00570

39. Schleich N., Sibret P., Danhier P., Ucakar B., Laurent S., Muller R. N., Danhier F. Dual anticancer drug/superparamagnetic iron oxide-loaded PLGA-based nanoparticles for cancer therapyand magnetic resonance imaging. Inter. J.Pharmac. 2013, 447 (1-2), 94-101.

40. Zuo H. D., Yao W. W., Chen T. W., Zhu J., Zhang J. J,, Pu Y., Zhang X. M. The Effect of Superparamagnetic Iron Oxide with iRGD Peptide on the Labeling of Pancreatic Cancer CellsIn Vitro: A Preliminary Study. BioMed Res. Inter. 2014, P. 1-8. https:// doi.org/10.1155/2014/852352

41. Huh Y. M., Jun Y. W., Song H. T., Kim S., Choi J. S., Lee J. H., Yoon S., Kim K. S., Shin J. S., Suh J. S., Cheon J. In vivo magnetic resonance detection of cancer by using multifunctional magnetic nanocrystals. J. Am. Chem. Soc. 2005, 127 (35), https://doi. org/10.1021/ja052337c

42. Oghabian M. A., Jeddi-Tehrani M., Zolfaghari A., Shamsipour F., Khoei S., Amanpour S. Detectability of Her2 positive tumors using monoclonal antibody conjugated iron oxide nanoparticles in MRI. J. Nanosci. Nanotechnol. 2011, 11 (6), 5340-5344. https://doi.org/ 10.1166/jnn.2011.3775

43. Marchesan S., Kostarelos K., Bianco A., Prato M. The winding road for carbon nanotubes in nanomedicine. Mater. Today. 2015, V. 18, P. 12-19. https://doi.org/10.1016/j-mattod.2014.07.009

44. Lacerda L., Bianco A., Prato M., Kostarelos K. Carbon nanotubes as nanomedicines: From toxicology to pharmacology. Adv. Drug Deliv. Rev. 2006, V. 58, P. 1460-1470. https://doi. org/10.1016/j.addr.2006.09.015

45. Siu K. S., Chen D., Zheng X., Zhang X., Johnston N., Liu Y., Yuan K., Koropatnick J., Gillies E. R., Min W. P. Non-covalently f unctionalized single-walled carbon nanotube for topical siRNA delivery into melanoma. Biomaterials. 2014, V. 35, P. 3435-3442. https://doi.org/10.1016/j. biomaterials.2013.12.079

46. Sanginario A., Miccoli B., Demarchi D. Carbon Nanotubes as an Effective Opportunity for Cancer Diagnosis and Treatment. Biosensors. 2017, 7 (4), 9. https://doi.org/ 10.3390/ bios7010009.

47. Zununi Vahed S., Salehi R., Davaran S., Sharifi S. Liposome-based drug co-delivery systems in cancer cells. Mater. Sc. Engin: C. 2017, V. 71, P. 1327-1341. https://doi. org/10.1016/j.msec.2016.11.073

48. Eloy J. O., Petrilli R., Topan J. F., Antonio H. M., Barcellos J. P., Chesca D. L., Serafini L. N, Tiezzi D. G, Lee R. J., Marchet-ti J. M. Co-loaded paclitaxel/rapamycin liposomes: development, characterization and in vitro and in vivo evaluation for breast cancer therapy, Colloids Surf. B. Biointerfaces. 2016, V. 141, P. 74-82. https://doi.org/ 10.1016/j.colsurfb.2016.01.032

49. Elnakat H., Ratnam M. Distribution, functionality and gene regulation of folate receptor isoforms: implications in targeted therapy. Adv. Drug Deliv. Rev. 2004, V. 56, P. 1067-1084. https://doi.org/ 10.1016/j. addr.2004.01.001

50. Chaudhury A., Das S. Folate receptor targeted liposomes encapsulating anti-cancer drugs, Curr. Pharm. Biotechnol. 2015, V. 16, P. 333-343 https://doi.org/ 10.2174/13892 01016666150118135107

51. Wu D, Zheng Y., Hu X., Fan Z, Jing X. Anti-tumor activity of folate targeted biodegradable polymer-paclitaxel conjugate micelles on EMT-6 breast cancer model. Mater. Sci. Eng. C. 2015, V. 53, P. 68-75. https://doi. org/10.1016/j.msec.2015.04.012

52. Yang T., Li B., Qi S., Liu Y., Gai Y., Ye P., Yang G., Zhang W., Zhang P., He X., Li W., Zhang Z., Xiang G., Xu C. Co-delivery of doxorubicin and Bmi1 siRNA by folate receptor targeted liposomes exhibits enhanced anti-tumor effects in vitro and in vivo. Theranostics. 2014, V. 4, P. 1096-1111. https://doi.org/ 10.7150/thno.9423

53. Peng Z., Wang C., Fang E., Lu X., Wang G., Tong Q. Co-delivery of doxorubicin and SATB1 shRNA by thermosensitive magnetic cationic liposomes for gastric cancer therapy. PLoS One. 2014, V. 9, P. e92924. https:// doi.org/ 10.1371/journal.pone.0092924

54. Connelly C. M., Uprety R., Hemphill J., Deiters A. Spatiotemporal control of microRNA function using light-activated antagomirs, Mol. BioSyst. 2012, V. 8, P. 2987-2993. https://doi.org/10.1039/c2mb25175b

55. Riaz M., Riaz M., Zhang X., Lin C., Wong K, Chen X. Yang Z. Surface Functionalization and Targeting Strategies of Liposomes in Solid Tumor Therapy: A Review. Inter. J. Mol. Sci. 2018, 19 (1), 195. https://doi.org/ 10.3390/ijms19010195

56. Biswas S., Kumari P., Lakhani P. M., Ghosh B. Recent advances in polymeric micelles for anti-cancer drug delivery. Europ. J. Pharmac. Sc. 2016, V. 83, P. 184-202. https://doi. org/10.1016/j.ejps.2015.12.031

57. Lohcharoenkal W., Wang L, Chen Y. C., Rojanasakul Y. Protein Nanoparticles as Drug Delivery Carriers for Cancer Therapy. BioMed Res. Inter. 2014, P. 1-12. https:// doi.org/10.1155/2014/180549

58. Weber C., Coester C., Kreuter J., Langer K. Desolvation process and surface characterisation of protein nanoparticles. Inter. J. Pharmac. 2000, 194 (1), 91-102. https://doi.org/

59. Teng Z., Luo Y., Wang T., Zhang B., Wang Q. Development and application of nanoparticles synthesized with folic acid conjugated soy protein. J. Agricult. Food Chemy. 2013, V. 61, P. 2556-2564. https://doi.org/10.1021/jf4001567

60. Gulf am M., Kim J., Lee J. M., Ku B., Chung B. H., Chung B. G. Anticancer drug-loaded gliadin nanoparticles induced apoptosis in breast cancer cells. Langmuir. 2012, V. 28, P. 8216- 8223. https://doi. org/10.1021/la300691n

61. Elzoghby A. O., Saad N. I., Helmy M. W., Samy W. M., Elgindy N. A. Ionically-crosslinked milk protein nanoparticles as flutamide carriers for effective anticancer activity in prostate cancer-bearing rats. Europ. J. Pharmac. Biopharmac. 2013, 85 (3), part A, 444-451. https://doi.org/10.1016Zj.ejpb.2013.07.003

62. Bazak R., Houri M., El Achy S., Kamel S., Refaat T. Cancer active targeting by nanoparticles: a comprehensive review of literature. J. Cancer Res. Clin. Oncol. 2014, 141 (5), 769-784. https://doi.org/ 10.1007/ s00432-014-1767-3

63. Eigenbrot C., Ultsch M., Dubnovitsky A., Abrahmsen L., Hard T. Structural basis for high-affinity HER2 receptor binding by an engineered protein. Proc. Nat. Acad, Sci. USA. 2010, 107 (34), 15039-15044. https:// doi.org/10.1073/pnas.1005025107

64. Zhang J. M., Zhao X. M., Wang S. J., Ren X. C., Wang N., Han J.-Y., Jia L. Z. Evaluation of 99mTc peptide ZHER2: 342Affibody®molecule forin vivomolecular imaging. The Brit. J. Radiol. 2014, 87 (1033), 20130484.

65. Ghanemi M., Pourshohod A., Ghaffari M. A., Kheirollah A., Amin M., Zeinali M., Jama-lan M. Specific Targeting of HER2-Positive Head and Neck Squamous Cell Carcinoma Line HN5 by Idarubicin ZHER2 Affibody Conjugate. Curr. Cancer Drug Targets. 2019, 19 (1), 65-73. https://doi.org/ 10.2174/156 8009617666170427105417

66. Glazer E. S., Massey K. L., Zhu C., Curley S. A. Pancreatic carcinoma cells are susceptible to noninvasive radio frequency fields after treatment with targeted gold nanoparticles. Surgery. 2010, 148 (2), 319-324. https://doi.org/ https://doi.org/10.1016/j.surg.2010.04.025

67. Talekar M., Kendall J., Denny W., Garg S. Targeting of nano- particles in cancer: drug

delivery and diagnostics. Anticancer Drugs. 2011, 22 (10), 949-962. https://doi.org/ 10.1097/CAD.0b013e32834a4554

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

68. Wang Z, Gu F,, Zhang L., Chan J. M., Radovic-Moreno A., Shaikh M. R. Biofunctionalized targeted nanoparticles for thera- peutic applications. Expert Opin. Biol. Ther. 2008, 8 (8), 1063-1070. https://doi.org/ 10.1517/14712598.8.8.1063

69. Lebel M.-E., Chartrand K,, Tarrab E., Savard P., Leclerc D., Lamarre A. Potentiating Cancer Immunotherapy Using Papaya Mosaic Virus-Derived Nanoparticles. Nano Letters.

2016, 16 (3), 1826-1832. https://doi.org/ 10.1021/acs.nanolett.5b04877.

70. Jinu U., Gomathi M., Saiqa I., Geetha N., Benelli G., Venkatachalam P. Green engineered biomolecule-capped silver and copper nanohybrids using Prosopis cineraria leaf extract: Enhanced antibacterial activity against microbial pathogens of public health relevance and cytotoxicity on human breast cancer cells (MCF-7). Microbial Pathogenesis.

2017, V. 105, P. 86-95. https://doi. org/10.1016/j.micpath.2017.02.019

71. Joshi M. D., Patravale V., Prabhu R. Polymeric nanoparticles for targeted treatment in oncology: current insights. Inter. J. Nanomed. 2015, P. 1001. https:// doi.org/ 10.2147/IJN.S56932

72. Torchilin V. Tumor delivery of macromolecular drugs based on EPR effect. Adv. Drug Delivery Rev. 2011, V. 63, P. 131-135. https://doi. org/10.1016/j.addr.2010.03.011

73. Greish K. In Cancer Nanotechnology: Methods and Protocols. Ed. R. S. Grobmyer and M. B. Moudgil. Humana Press, Totowa, NJ. 2010, 25-37.

74. Maeda H. Nakamura, Fang J. The EPR effect for macromolecular delivery to solid tumors: improvement of tumor uptake lowering of systemic toxicity and distinct tumor imaging in vivo. Adv. Drug Deliv. Rev. 2013, V. 65, P. 71-79. https://doi.org/10.1016/j. addr.2012.10.002

75. Sun C., Lee J. S., Zhang M. Magnetic nanoparticles in MR imaging and drug delivery. Adv. Drug Deliv. Rev. 2008, V. 60, P. 1252-1265. https://doi.org/ 10.1016/j.addr.2008.03.018

76. Aires A., Ocampo S. M., Simoes B. M., Rodriguez M. J., Cadenas J. F., Couleaud P., Spence K., Latorre A., Miranda R., Somoza A., Clarke R. B., Carrascosa J. L., Cortajarena A. L. Multif unctionalized iro n isE_pj oxide nanoparticles for selective drug delivery to CD44-positive ^cancer cells. Nanotechnol. 2016, V. 27, P. 065103.

77. Marciello M., Pellico J., Fernandez-Barahona I., Herranz F., Ruiz-Cabello J., Filice M. Recent advances in the preparation and application of multifunctional iron oxide and liposome-

based nanosystems for multimodal diagnosis and therapy. Interface Focus. 2016, 6 (6), 2016005 5. https://doi.org/10.1098/ rsfs.2016.0055

78. Guo J., Rahme K, He Y., Li L.-L., Holmes J., O'Driscoll C. Gold nanoparticles enlighten the future of cancer theranostics. Inter. J. Nanomed. 2017, V. 12, P. 6131-6152. https://doi.org/10.2147/IJN.S140772

79. Jin Y. Multifunctional compact hybrid Au nanoshells: a new generation of nanoplasmonic probes for biosensing, imaging, and controlled release. Acc. Chem. Res. 2014, 47 (1), 138-148. https://doi. org/10.1021/ar400086e

80. Zhao Y., Pang B., Luehmann H. Gold nanoparticles doped with (199) Au atoms and their use for targeted cancer imaging by SPECT. Adv. Healthc. Mater. 2016, 5 (8), 928-935. https://doi.org/10.1002/ adhm.201500992

81. Liu J., Zhang L., Lei J., Ju H. MicroRNA-Responsive Cancer Cell Imaging and Therapy

with Functionalized Gold Nanoprobe. ACS Appl. Mater. Interfaces. 2015, 7 (34), 19016-19023. https://doi.org/10.1021/ acsami.5b06206

82. Li K., Nejadnik H., Daldrup-Link H. E. Next-generation superparamagnetic iron oxide nanoparticles for cancer theranostics. Drug Discov. Today. 2017, 22 (9), 1421-1429. https://doi.org/ 10.1016/j.drudis.2017.04.008

83. Daldrup-Link H. E. Mohanty S., Ansari C., Lenkov O., ShawF., Ito K., Hong S. H., Hoffmann M., Pisani L., Boudreau N., Gambhir S. S., Coussens L. M. Alk5 inhibition increases delivery of macromolecular and protein-bound contrast agents to tumors. JCI Insight. 2016, V. 1, P. e85608. https://doi. org/ 10.1172/jci.insight.85608

84. Zaimy M. A., Saffarzadeh N., Mohammadi A., Pourghadamyari H., Izadi P., Sarli A., Tavakkoly-Bazzaz J. New methods in the diagnosis of cancer and gene therapy of cancer based on nanoparticles. Cancer Gene Ther. 2017, 24 (6), 233-243.

Д1АГНОСТИКА, В1ЗУАЛ1ЗАЦ1Я ТА Л1КУВАННЯ РАКУ З ВИКОРИСТАННЯМ НАНОРОЗМ1РНИХ СТРУКТУР

О. О. Озкан, М. Карахан

Ушверситет Ускюдар, Стамбул, Туреччина

E-mail: karahan@uskudar.edu.tr

Сучасш дослщження спрямоваш на по-шук нових стратегш лшування раку, як не мають значних побiчних ефекйв i б^ьш ефективн порiвняно з традицшними методами, включаючи xipypri4^ втручання, променеву тератю i хiмiотерапiю.Застосу-вання нанорозмiрних структур уможливлюе використання новггшх пiдходiв для лшування раку. Системи наночастинок тдвищують ефектившсть терапевтичних препарайв, знижуючи 1х побiчнi ефекти. Хоча в багатьох дослщженнях повщомляеться про розробку нових методiв лшування ракових клгтин в майбутньому, клШчний устх не дозволяе зрозумiти вплив типу, розмiру та дози на-ночасток на област 1х застосування. Таким чином, в цьому оглядi проiлюстровано мож-ливiсть використання наночасток в дiагнос-тицi, вiзуалiзацii та лiкуваннi раку.

Ключовi слова: дiагностика раку, вiзуалi-зацiя та лiкування, наночастинки.

ДИАГНОСТИКА, ВИЗУАЛИЗАЦИЯ И ЛЕЧЕНИЕ РАКА С ИСПОЛЬЗОВАНИЕМ НАНОРАЗМЕРНЫХ СТРУКТУР

О. О. Озкан, М. Карахан

Университет Ускюдар, Стамбул, Турция

E-mail: karahan@uskudar.edu.tr

Современные исследования направлены на поиск новых стратегий лечения рака, не имеющих значительных побочных эффектов и более эффективных по сравнению с традиционными методами, включая хирургическое вмешательство, лучевую терапию и химиотерапию. Применение наноразмерных структур дает возможность использовать полезные подходы для лечения рака.Системы наночастиц повышают эффективность терапевтических препаратов, снижая их побочные эффекты. Хотя во многих исследованиях сообщается о разработке новых методов лечения раковых клеток в будущем, клинический успех не позволяет понять влияние типа, размера и дозы наночастиц на области их применения. Таким образом, в настоящем обзоре проиллюстрирована возможность использования наночастиц в диагностике, визуализации и лечении рака.

Ключевые слова: диагностика рака, визуализация и лечение, наночастицы.

i Надоели баннеры? Вы всегда можете отключить рекламу.