Вы находитесь на странице: 1из 12

Whole-cell SELEX aptamer-functionalised poly(ethyleneglycol)-

poly(ε-caprolactone) nanoparticles for enhanced targeted glioblastoma


therapy

Glioma was the most commonly diagnosed brain cancer at the time,
accounting for approximately 45%e50% of primary brain cancer cases [2]

Among all the types of cancer glioma was one of the most commonly diagnosed
brain cancer ,normally 45% to 50% cases were of glioma.[2]

. The poor prognosis associated with GBM is largely because of the poorly
defined tumour border of GBM, incomplete resection of the tumour and
tumour recurrence after surgical resection [4]. Chemotherapy is an
essential part of GBM therapy, but it is mainly ineffective due to the
small amount of drug accumulation in GBM cells, drug resistance and
serious side effects [4]

For the treatment of glioma chemotherapy is normally done but there are
many problems assosiated with chemotherapy includes ineffectiveness due
to low ammount of drug accumulation in GBM cells , many side effects and
drug resistance.[4]

There are several targeting strategies including receptor-mediated,


carrier-mediated, adsportive-mediated targeting and other physical
methods [5].

There are a number of targeting schemes which includes


adsportive-mediated ,receptor-mediated,carrier-mediated targeting and
other physical strategies.[5]

Aptamers are single stranded DNA, RNA or modified nucleic acids that can
specifically bind to their targets [6].

By definition aptamers are single stranded DNA RNA or altered nucliec


acids which have the ability to bind with a particular target.[6]

Specific aptamers can be selected from a library of random sequences


through systematic evolution of ligands by exponential enrichment (SELEX)
[6,7].
Particular aptamers can be chosen from a library of random sequence with
the help of systematic evolution of ligands by exponential enrichment
(SELEX)[6,7]

Docetaxel (DTX), a taxane derivative similar to paclitaxel, is an


inhibitor of microtubule depolymerisation [8] that is widely used for the
treatment of many cancers including brain cancer [9].

GMT8 aptamers were synthesised by the Sangon Biotech Co., Ltd (Shanghai,
China). Methoxy poly(ethyleneglycol)-poly(ε-caprolactone) (MPEG-PCL,
Mw: 3k15k) and R-carboxyl poly(ethylene glycol)-poly(ε-caprolactone)
(HOOC-PEG-PCL, Mw: 3.4k-15k) were synthesised as previously described
[10].

Targeted delivery by systems functionalised with specific ligands is a


promising strategy for the carriers, as they could have both the
anti-cancer property of the drugs and the active targeting effect of the
ligand at the same time. Several strategies were developed for this
purpose, which has shown promising results [26e28].

Carriers are the main components of targeted delivery systems. Many


carriers have been developed for drug delivery, including liposomes,
micelles, nanoparticles, polymersomes and dendrimers [29e33].

Patient safety, which is what researchers unceasingly aspire to achieve,


is a critical requirement for carriers [34].

Targeting ligands are the most important part of a targeted drug delivery
system. Many researchers focus on the newly emerged ligands for specific
receptors that are overexpressed in the target cells or on the redesign
of existing ligands, such as lactoferrin [36], angiopep-2 derived from
aprotinin [36], a 16-residue peptide (CDX) derived from candoxin [31] and
a 30-amino-acid peptide (leptin30) derived from an endogenic
hormone-leptin [37].

However, these ligands, mostly proteins or peptides, are related to


immunogenicity and competition with endogenous ligands, which may reduce
their targeting effect. Several methods have been established to select
better targeting ligands, including SELEX and phage display [6,38].

These methods can select specific short sequences of peptides or DNA from
a library that rules out the endogenous effect and immunogenicity; RGD
was the most successful example that has been demonstrated by several
groups [19,39e42].

. GMT8 is a DNA aptamer selected from a library by whole-cell SELEX against


glioma cells [7]. GMT8 aptamers can bind with high specificity to U87 cells,
which is consistent with our results showing that GMT8 aptamers
significantly enhance the uptake of ApNP compared with unmodified NP by
U87 cells. These results suggest that GMT8 aptamers could serve as an
effective ligand for GBM therapy.[article]

We established a GBM-targeted delivery system utilising GMT8 aptamers as


the targeting ligand. In vitro cellular uptake and tumour spheroid uptake
demonstrated the target effect of ApNP; cell apoptosis and tumour spheroid
growth inhibition attested to its improved anti-tumour cell growth effect.
In vivo imaging and antiGBM experiments further demonstrated that ApNP
could effectively target GBM and have an anti-GBM effect.[article]

Reference

[2]
Guo J, Gao X, Su L, Xia H, Gu G, Pang Z, et al. Aptamer-functionalized
PEGPLGA nanoparticles for enhanced anti-glioma drug delivery.
Biomaterials 2011;32(31):8010e20.

[4]

Lawson HC, Sampath P, Bohan E, Park MC, Hussain N, Olivi A, et al.


Interstitial chemotherapy for malignant gliomas: the Johns Hopkins
experience. J Neurooncol 2007;83(1):61e70.

[5] Ren J, Shen S, Wang D, Xi Z, Guo L, Pang Z, et al. The targeted delivery
of anticancer drugs to brain glioma by PEGylated oxidized multi-walled
carbon nanotubes modified with angiopep-2. Biomaterials
2012;33(11):3324e33.

[6] Shangguan D, Li Y, Tang Z, Cao ZC, Chen HW, Mallikaratchy P, et al.


Aptamers evolved from live cells as effective molecular probes for cancer
study. Proc Natl Acad Sci U S A 2006;103(32):11838e43.

[7] Bayrac AT, Sefah K, Parekh P, Bayrac C, Gulbakan B, Oktem HA, et al.
In vitro Selection of DNA aptamers to glioblastoma Multiforme. ACS Chem
Neurosci 2011;2(3):175e81.

[9] Sampath P, Rhines LD, DiMeco F, Tyler BM, Park MC, Brem H. Interstitial
docetaxel (taxotere), carmustine and combined interstitial therapy: a
novel treatment for experimental malignant glioma. J Neurooncol
2006;80(1):9e17.

[10] Pang Z, Lu W, Gao H, Hu K, Chen J, Zhang C, et al. Preparation and


brain delivery property of biodegradable polymersomes conjugated with
OX26. J Control Release 2008;128(2):120e7.

[26] Li J, Gu B, Meng Q, Yan Z, Gao H, Chen X, et al. The use of myristic


acid as a ligand of polyethylenimine/DNA nanoparticles for targeted gene
therapy of glioblastoma. Nanotechnology 2011;22(43):435101.

[27] Pang Z, Feng L, Hua R, Chen J, Gao H, Pan S, et al.


Lactoferrin-conjugated biodegradable polymersome holding doxorubicin
and tetrandrine for chemotherapy of glioma rats. Mol Pharm
2010;7(6):1995e2005.

[28] Pang Z, Gao H, Yu Y, Guo L, Chen J, Pan S, et al. Enhanced


intracellular delivery and chemotherapy for glioma rats by
transferrin-conjugated biodegradable polymersomes loaded with
Doxorubicin. Bioconjug Chem 2011;22(6): 1171e80.
[29] Pardridge WM. Vector-mediated drug delivery to the brain. Adv Drug
Deliv Rev 1999;36(2e3):299e321.

[30] Tang R, Ji W, Panus D, Palumbo RN, Wang C. Block copolymer micelles


with acid-labile ortho ester side-chains: Synthesis, characterization,
and enhanced drug delivery to human glioma cells. J Control Release
2011;151(1):18e27.

[31] Zhan C, Li B, Hu L, Wei X, Feng L, Fu W, et al. Micelle-based


brain-targeted drug delivery enabled by a nicotine acetylcholine receptor
ligand. Angew Chem Int Ed Engl; 2011.

[32] Gao X, Wu B, Zhang Q, Chen J, Zhu J, Zhang W, et al. Brain delivery


of vasoactive intestinal peptide enhanced with the nanoparticles
conjugated with wheat germ agglutinin following intranasal
administration. J Control Release 2007;121(3):156e67.

[33] Han L, Huang R, Li J, Liu S, Huang S, Jiang C. Plasmid pORF-hTRAIL


and doxorubicin co-delivery targeting to tumor using peptide-conjugated
polyamidoamine dendrimer. Biomaterials 2011;32(4):1242e52.

[34] Kim SY, Lee YM, Baik DJ, Kang JS. Toxic characteristics of methoxy
poly (ethylene glycol)/poly(epsilon-caprolactone) nanospheres; in vitro
and in vivo studies in the normal mice. Biomaterials 2003;24(1):55e63.

[37] Liu Y, Li J, Shao K, Huang R, Ye L, Lou J, et al. A leptin derived


30-amino-acid peptide modified pegylated poly-L-lysine dendrigraft for
brain targeted gene delivery. Biomaterials 2010;31(19):5246e57.

[38] Pasqualini R, Ruoslahti E. Organ targeting in vivo using phage


display peptide libraries. Nature 1996;380(6572):364e6.

[42] Tyler MA, Ulasov IV, Borovjagin A, Sonabend AM, Khramtsov A, Han Y,
et al. Enhanced transduction of malignant glioma with a double targeted
Ad5/3- RGD fiber-modified adenovirus. Mol Cancer Ther 2006;5(9):2408e16.

[19] Zhan C, Gu B, Xie C, Li J, Liu Y, Lu W. Cyclic RGD conjugated


poly(ethylene glycol)-co-poly(lactic acid) micelle enhances paclitaxel
anti-glioblastoma effect. J Control Release 2010;143(1):136e42
Activatable Fluorescence/MRI Bimodal Platform for Tumor Cell Imaging via
MnO2 Nanosheet−Aptamer Nanoprobe

We report a novel dual-activatable fluorescence/MRI strategy for bimodal


cancer imaging using a nanoprobe based on the physisorption of Cy5-labeled
aptamers on redoxable MnO2 nanosheets (Scheme 1). To date, various
materials based on Mn, such as MnCl2, 12 manganese chelates,13 MnO nano
particles,14 and hybrid nanomaterials,15 have been used as contrast
agents.

A novel dual-activatable fluorescence/MRI bimodal platform is designed


for tumor cell imaging by using a redoxable manganese dioxide (MnO2)
nanosheet− aptamer nanoprobe. The redoxable MnO2 nanosheet acts as a DNA
nanocarrier, fluorescence quencher, and intracellular glutathione
(GSH)-activated MRI contrast agent. In the absence of target cells,
neither fluorescence signaling nor MRI contrast of the nanoprobe is
activated. In the presence of target cells, the binding of aptamers to
their targets weakens the adsorption of aptamers on the MnO2 nanosheets,
causing partial fluorescence recovery, illuminating the target cells, and
also facilitating the endocytosis of nanoprobes into target cells. After
endocytosis, the reduction of MnO2 nanosheets by GSH further activates
the fluorescence signals and generates large amounts of Mn2+ ions suitable
for MRI. This platform should facilitate the development of various
dualactivatable fluorescence/MRI bimodalities for use in cells or in
vivo.[16]

Reference

[12]Aoki, I.; Takahashi, Y.; Chuang, K. H.; Silva, A. C.; Igarashi, T.;
Tanaka, C.; Childs, R. W.; Koretsky, A. P. NMR Biomed. 2006, 19, 50.

[13] (a) Troughton, J. S.; Greenfield, M. T.; Greenwood, J. M.; Dumas,


S.; Wiethoff, A. J.; Wang, J.; Spiller, M.; McMurry, T. J.; Caravan, P.
Inorg. Chem. 2004, 43, 6313. (b) Loving, G. S.; Mukherjee, S.; Caravan,
P. J. Am. Chem. Soc. 2013, 135, 4620.

[14] Kim, T.; Momin, E.; Choi, J.; Yuan, K.; Zaidi, H.; Kim, J.; Park,
M.; Lee, N.; McMahon, M. T.; Quinones-Hinojosa, A.; Bulte, J. W.; Hyeon,
T.; Gilad, A. A. J. Am. Chem. Soc. 2011, 133, 2955

[15] Schladt, T. D.; Shukoor, M. I.; Schneider, K.; Tahir, M. N.; Natalio,
F.; Ament, I.; Becker, J.; Jochum, F. D.; Weber, S.; Kö hler, O.; Theato,
P.; Schreiber, L. M.; Sö nnichsen, C.; Schrö der, H. C.; Müller, W.
E.; Tremel, W. Angew. Chem., Int. Ed. 2010, 49, 3976

[16] Activatable Fluorescence/MRI Bimodal Platform for Tumor Cell Imaging


via MnO2 Nanosheet−Aptamer Nanoprobe
We described the development of integrin-targeting magnetic
nanoparticles (Aptavb3-MNPs) by modifying integrin avb3-targeting
aptamer (Aptavb3) to produce a particle with high ability to detect
integrin avb3 expression in cancer cells during angiogenesis using MR
imaging. Aptavb3-MNPs exhibited no cytotoxicity and excellent
tumor-targeting detection ability with high magnetic sensitivity,
thereby demonstrating the superb performance of Aptavb3 as a targeting
vector. Furthermore, the incorporation of other diagnostic agents and
therapeutic agents in Aptavb3-MNPs might provide more accurate tumor
diagnosis and therapy.[1]

Refernce

[1] Aptamer-conjugated magnetic nanoparticles enable efficient targeted


detection of integrin avb3 via magnetic resonance imaging

MUC1 aptamer conjugated to chitosan nanoparticles, an efficient targeted


carrier designed for anticancer SN38 delivery
Aptamer-Conjugated Nanoparticles for Cancer Cell Detection

The development of aptamers for molecular recognition addresses several


of these issues: aptamers possess a low molecular weight, exhibit easy
and reproducible synthesis, easy modification,6 fast tissue penetration,
low toxicity or immunogenicity,7,8 easy storage, and high binding
affinity and selectivity.9,10

The panel of aptamers generated is ideal for profiling and personalized


medicine, as different aptamers recognize different molecular targets on
the surface of the cell which could be subsequently be correlated to
advancing patient and treatment outcomes. Panels of aptamers targeting
leukemia cell lines11, lung cancer cell lines12, liver cancer cell lines13,
and lymphoma cell lines14 have previously been described.

Reference

6. Li J, Fang XH, Tan W. Biochem Biophys Res Commun. 2002; 292:31–40.


[PubMed: 11890667]

7. Knox KK, Brewer JH, Henry JM, Harrington DJ, Carrigan DR. Clin
Infectious Diseases. 2000; 31(4):894–903.

8. Cerchia L, Hamm J, Libri D, Tavitian B, Franciscis V. FEBS Letters.


2002; 528:12–16. [PubMed: 12297271]

9. Gold L. J Biol Chem. 1995; 270:13581–13584. [PubMed: 7775406]

10. Hermann T, Patel D. J Science. 2000; 287:820–825.

11. Shangguan D, Li Y, Tang Z, Cao ZC, Mallikaratchy P, Sefah K, Yang CJ,


Tan W. Proc Natl Acad Sci USA. 2006; 103:11838. [PubMed: 16873550]

12. Chen H, Medley CD, Sefah K, Shangguan D, Tang Z, Smith JE, Meng L,
Tan W. Chem Med Chem. 2008; 3:991. [PubMed: 18338423]

13. Shangguan D, Meng L, Cao ZC, Xiao Z, Fang X, Li Y, Cardona D, Witek


RP, Liu C, Tan W. Anal Chem. 2008; 80:721. [PubMed: 18177018]

14. Tang Z, Shangguan D, Wang K, Shi H, Sefah K, Mallikaratchy P, Chen


WH, Li Y, Tan W. Anal Chem. 2007; 79:4900. [PubMed: 17530817]..
Aptamer-conjugated nanomaterials and their applications

By combining the inherent features of nanomaterials with the specific


recognition ability of aptamers, a range of nanomaterial- aptamer
conjugates have proven their utility in multiple areas. 6,7

Aptamers are single-stranded oligonucleotides, DNA or RNA, with the


ability to bind to non-nucleic acid target molecules, such as peptides,
proteins, drugs, organic and inorganic molecules, or even whole cells,
with high affinity and specificity.8-12 They are isolated and chemically
synthesized from 1012 –1015 combinatorial oligonucleotide libraries by
a process known as in vitro systematic evolution of ligands by exponential
enrichment (SELEX). 13,14 Over multiple rounds of selection (generally
6–18 rounds), quite large populations (>1013 different sequences) can
be sieved, and the few nucleic acid species with specificity to the target
can be isolated[15]

Reference

6. Wang H, Yang RH, Yang L, Tan WH. ACS Nano. 2009; 3:2451–2460. [PubMed:
19658387]

7. Stadler N, Chi C, Lelie DVD, Gang O. Nanomed. 2010; 5:319–334.

8. Mairal T, Özalp VC, Sánchez PL, Mir M, Katakis I, O'Sullivan CK. Anal
Bioanal Chem. 2008; 390:989–1007. [PubMed: 17581746]

11. Bunka DHJ, Stockley PG. Nature Reviews Microbiol. 2006; 4:588–596.
12. Sefah K, Tang ZW, Shangguan DH, Chen H, Lopez-Colon D, Li Y, Parekh
P, Martin J, Meng L, Phillips JA, Kim YM, Tan WH. Leukemia. 2009; 23:235–
244. [PubMed: 19151784]

13. Wilson DS, Szostak JW. Annu Rev Biochem. 1999; 68:611–647. [PubMed:
10872462]

14. Ellington D, Szostak JW. Nature. 1990; 346:818–822. [PubMed:


1697402]

Вам также может понравиться