ArticlePDF AvailableLiterature Review

Targeted Delivery of Small Interfering RNA: Approaching Effective Cancer Therapies

Authors:

Abstract

Three of the primary requirements for the development of effective dual-targeting therapeutic modalities for the treatment of cancer are the tumor-targeted delivery of the therapeutic molecules of interest to the tumor site(s) in the body (both primary and metastatic), passage of the molecular therapeutic through the cell membrane, and targeting specifically a growth or apoptotic pathway. However, lack of efficient targeted delivery, low transfection efficiency, instability to nucleases, poor tissue penetration, and nonspecific immune stimulation have hindered the translation of small interfering RNA (siRNA) into clinical applications. The development of a systemically administered, tumor-specific immunoliposome nanocomplex with high transfection efficiency could overcome these limitations and thus realize the potential of siRNAs to become effective anticancer clinical modalities.
2008;68:1247-1250. Cancer Res
Kathleen F. Pirollo and Esther H. Chang
Effective Cancer Therapies
Targeted Delivery of Small Interfering RNA: Approaching
Updated version
http://cancerres.aacrjournals.org/content/68/5/1247
Access the most recent version of this article at:
Cited Articles
http://cancerres.aacrjournals.org/content/68/5/1247.full.html#ref-list-1
This article cites by 27 articles, 9 of which you can access for free at:
Citing articles
http://cancerres.aacrjournals.org/content/68/5/1247.full.html#related-urls
This article has been cited by 4 HighWire-hosted articles. Access the articles at:
E-mail alerts
related to this article or journal.Sign up to receive free email-alerts
Subscriptions
Reprints and
.pubs@aacr.orgDepartment at
To order reprints of this article or to subscribe to the journal, contact the AACR Publications
Permissions
.permissions@aacr.orgDepartment at
To request permission to re-use all or part of this article, contact the AACR Publications
Research.
on June 11, 2013. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from
Targeted Delivery of Small Interfering RNA:
Approaching Effective Cancer Therapies
Kathleen F. Pirollo and Esther H. Chang
Department of Oncology, Georgetown University Medical Center, Washington, District of Columbia
Abstract
Three of the primary requirements for the development of
effective dual-targeting therapeutic modalities for the treat-
ment of cancer are the tumor-targeted delivery of the
therapeutic molecules of interest to the tumor site(s) in the
body (both primary and metastatic), passage of the molecular
therapeutic through the cell membrane, and targeting
specifically a growth or apoptotic pathway. However, lack of
efficient targeted delivery, low transfection efficiency, insta-
bility to nucleases, poor tissue penetration, and nonspecific
immune stimulation have hindered the translation of small
interfering RNA (siRNA) into clinical applications. The
development of a systemically administered, tumor-specific
immunoliposome nanocomplex with high transfection effi-
ciency could overcome these limitations and thus realize the
potential of siRNAs to become effective anticancer clinical
modalities. [Cancer Res 2008;68(5):1247–50]
Although the use of potent, sequence-specific small interfering
RNAs (siRNA) to suppress expression of specific transcripts was
originally a useful technique for probing gene function in vitro,
their successful application in vivo in animal models against a
spectrum of diseases, including cancer (1–5), has spurred interest
in developing this approach for siRNA-based therapeutics.
However, there are still significant obstacles to be overcome before
these molecules can be used in the clinic as anticancer agents,
the focus of this minireview. Perhaps foremost among these is the
issue of delivery. The in vivo use of siRNAs effectively against
cancer hinges on the availability of a delivery vehicle that can be
systemically administered to reach both primary and metastatic
tumor cells. Moreover, because sufficient intact, functional siRNA
must be delivered into the target cell to reach an effective
intracellular concentration, and to limit potential side effects due
to randomized, general transfection of normal, nontarget tissues, it
is also crucial to develop means of directing such a siRNA delivery
vehicle specifically to the target cells.
Naked siRNAs, delivered into the bloodstream, even when
chemically modified, have extremely short half-lives (seconds to
minutes) due to renal clearance (because of their small size; ref. 6).
In addition, whereas most RNases are inactive against double-
stranded RNA (dsRNA), some serum RNases can degrade siRNA (6).
Cellular uptake of naked siRNA is also limited. To overcome some
of these challenges, siRNAs have been complexed to a variety of
nonviral lipids or protein carriers, including cholesterol, liposomes,
antibody protomer fusions, cyclodextrin nanoparticles, fusogenic
peptides, aptamers, biodegradable polylactide copolymers, and
polymers (4–7). Positively charged cationic liposomes and poly-
mers, such as polyethyleneimine, are currently the two major
carriers used to complex with negatively charged siRNA for
systemic delivery (3, 8). Although most of the reports in the
literature use delivery approaches that are not systemically
administered and/or specifically targeted to the tumor, there are
a few reports of targeted i.v. delivery of siRNA in animal models of
cancer.
The RGD peptide and transferrin (Tf), as well as antibodies
and antibody fragments [such as anti-Tf receptor (TfR) and anti–
epidermal growth factor receptor], have been used as targeting
ligands for i.v. siRNA delivery against tumors (reviewed in ref. 8).
Schiffelers et al. (9) linked siRNA against vascular endothelial
growth factor (VEGF) receptor 2 to polyethyleneimine that was
PEGylated with an RGD peptide ligand at the distal end as a
means to target tumor neovasculature. They reported inhibition
of both tumor angiogenesis and growth rate in mice bearing
murine neuroblastoma N2A tumor xenografts. Similarly, anti-
angiogenic effects were also observed in ocular neovasculariza-
tion in the herpes simplex virus disease model (10). Hu-
Lieskovan et al. (11), using a Tf-targeted, cyclodextrin-based
polycation for delivery of siRNA to tumors in a mouse model of
Ewing’s sarcoma, saw transient reduction of tumor growth.
Recently, Bartlett et al. (12) used a Tf-targeting,
64
Cu-labeled,
cyclodextrin-containing polycation to systemically deliver an anti-
luciferase siRNA molecule to Neuro2A-Luc tumor cells. Using
simultaneous positron emission tomography/computed tomogra-
phy to monitor siRNA whole-body biodistribution kinetics and
tumor localization to correlate biodistribution data with
functional efficacy, they concluded that the primary advantage
of the targeting molecule is associated with cellular uptake rather
than tumor localization. This same group had previously used Tf-
targeted polyplexes carrying an anti-luciferase siRNA to examine
the kinetics of siRNA-mediated silencing in mice bearing
Neuro2A-Luc tumors (13).
In a proof-of-principal study, Song et al. (14) reported that a
protamine-antibody fusion protein using the Fab fragment of HIV-1
envelope antibody (F105-P) as the targeting molecule i.v. delivered
FITC-labeled siRNA only to B-16 melanoma tumors modified to
express HIV env and not to normal tissues. They also used F105-P
to deliver a cocktail of siRNAs against c-myc, MDM-2, and VEGF to
mice bearing s.c. B-16 HIV env–expressing xenografts resulting in
tumor growth inhibition.
Recently, Pirollo et al. reported the development and use of a
tumor-specific, nanosized immunoliposome complex for systemic
delivery of siRNA (the scL delivery system; refs. 15–17). This
complex [TfR single-chain antibody fragment (TfRscFv)/liposome/
siRNA] is composed of an anti-HER-2 siRNA encapsulated by
a cationic [1,2-dioleoyl-trimethylammonium-propone (DOTAP)
Requests for reprints: Esther H. Chang, Department of Oncology, Georgetown
University Medical Center, TRB/E420, 3970 Reservoir Road Northwest, Washington,
DC 20057-1469. Phone: 202-687-8418; Fax: 202-687-8434; E-mail: change@
georgetown.edu.
I2008 American Association for Cancer Research.
doi:10.1158/0008-5472.CAN-07-5810
www.aacrjournals.org
1247
Cancer Res 2008; 68: (5). March 1, 2008
Review
Research.
on June 11, 2013. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from
dioleoyl phosphatidylethanolamine (DOPE)] liposome, the surface
of which is decorated with a targeting moiety, an anti-TfRscFv
(15–17). The TfRscFv contains the complete antibody binding site
for the epitope of the TfR recognized by the monoclonal antibody
(mAb) 5E9 (18, 19). TfR levels are elevated in various types of
cancer cells (20). Elevated TfR levels also correlate with the
aggressive or proliferative ability of tumor cells (20). The TfR
also recycles during internalization in rapidly dividing cells,
such as cancer cells (20), thus contributing to the uptake of
TfR-targeted nanocomplexes even in cancer cells where the
actual level of the TfR may not be elevated compared with
normal cells. Although we have also used Tf as a targeting ligand
(21), TfRscFv has certain advantages over the Tf molecule itself or
an entire mAb in targeting liposomes to cancer cells with elevated
TfR levels. (a) The size of the scFv (f28 kDa) is much smaller
than the Tf molecule (80 kDa) or the parental mAb (155 kDa).
The scFv-liposome-DNA complex may thus exhibit better
penetration into small capillaries characteristic of solid tumors.
(b) The smaller scFv has a practical advantage related to the
scaled-up production necessary for the clinical use. (c) Unlike Tf,
the scFv is a recombinant molecule and is not isolated from
blood.
To increase stability, the siRNA encapsulated in the immunoli-
posome complex in these studies was a modified hybrid form of
siRNA, a type of modification in which the double-stranded
molecule is composed of an unmodified antisense RNA strand and
a DNA sense strand that may or may not be modified. This
approach is different than the use of chemical modifications
(reviewed in ref. 8) to improve stability, reduce off-target effects,
and maintain efficacy of siRNAs. Three independent publications
(15, 22, 23) of RNA interference (RNAi) using these hybrid or
modified hybrid duplexes have reported that these antisense RNA/
sense DNA hybrid duplexes, which were called ‘‘siHybrids’’ (23),
were more potent (15, 22, 23) and led to longer-lasting (23) RNAi,
relative to corresponding unmodified siRNA. In contrast to the
trend for design of siRNA analogues wherein ‘‘more modification is
better,’’ the promising properties of siHybrids indicated that ‘‘less
may be more’’ (15). If so, this could translate into more cost-
effective RNAi by virtue of using a sense strand in which relatively
inexpensive DNA replaces more costly RNA having chemical
modifications. In addition, based on molecular appearance’’ to
Toll-like receptors in the innate immune system (24), a dsRNA/
DNA siHybrid might be less immunogenic than a homologous
dsRNA/RNA siRNA and may also induce less of an IFN response
than dsRNA.
One way to increase the efficacy of the siRNA after the
complex has reached the target cell is to enhance siRNA release
from the endosome. Thus, more of the siRNA is available in the
cytoplasm for knockdown of the target gene rather than being
trapped in the endosome and undergoing lysosomal enzymatic
degradation. Endosomal compartments are generally acidic in
nature. Various methods, including incorporation of pH-sensitive
components into liposomes, have been developed to enhance the
efficiency of liposomal payload delivery by exploiting this fact
(reviewed in ref. 25). The selective destabilization of liposomes
following acidification of the surrounding medium with resultant
release of the payload has been enhanced by inclusion of specific
lipids, many based on phosphatidylethanolamine or modifica-
tions thereof (e.g., DOPE). These undergo a lamellar to hexagonal
phase transition at low pH, releasing the liposomal contents.
Several pH-sensitive synthetic peptides have also been designed
in an attempt to produce peptides that can attach to, but not
perturb, the surface of a liposome at neutral pH and
subsequently fuse adjacent bilayers at acidic pH. Chen et al.
(26), as well as others (27), have designed linear and branched
histidine-lysine (HK) polymers of varying lengths. The histidine
component is believed to buffer and disrupt the endosomes.
The inclusion of such HK copolymers significantly increased
the transfection efficiency over cationic liposomes alone (24). To
further increase the efficacy of the scL complex by facilitating
endosomal release while maintaining the small size of the
nanoparticle complex, Pirollo et al. (17) also included a small
linear pH-sensitive peptide, HoKC {K[K(H)KKK]
5
K(H)KKC; adap-
ted from that of Aoki et al. (27)}, in the targeted immunolipo-
some complex. This HoKC peptide contains a cysteine residue at
the end, enabling it to be conjugated to the liposome through a
maleimide group.
The results published by Chang and colleagues (15–17) show,
using scanning probe microscopy, that this single-chain targeted
immunoliposome siRNA complex is a nanoparticle of uniform size,
even when the HoKC peptide is included. Their findings also show
that modifying the anti-HER-2 siRNA through use of a modified
DNA sequence as the sense strand significantly improved the in
vitro efficacy of the siRNA compared with standard duplex siRNA
and that this approach could significantly sensitize (by over 80-
fold) pancreatic cancer cells to the standard chemotherapeutic
agent gemcitabine.
The most significant findings of these reports are the tumor-
targeting in vivo results. When systemically (i.v. tail vein)
administered, both forms of the complex (with and without
inclusion of the HoKC peptide) delivered the fluorescently
labeled siRNA specifically and efficiently to tumors. This was
observed in both large primary prostate tumors (16) and in two
metastasis models using human pancreatic cancer and human
melanoma MDA435/LCC6 (16, 17). The ability of this approach
to efficiently target and transfect metastatic lesions is shown in
Fig. 1. The metastasis indicated by the arrow displays a high
level of fluorescence with no significant signal in the adjacent
normal lung tissue. Moreover, micrometastases near the larger
nodule (verified by histology) are also detectable, indicating that
even tiny nodules composed of only a few tumor cells can also
be reached and transfected via this complex. These results
confirm the tumor-targeting ability and the efficient delivery of
the siRNA by the nanocomplex containing the pH-sensitive
peptide.
More importantly, they were also able to show that this tumor-
specific delivery via the HoKC nanocomplex resulted not only in
virtually complete knockdown of HER-2 expression in the tumors
but also in concomitant changes in expression of pAKT, pMAPK,
Bcl-2 (down-modulation), and caspase-3 (up-regulation), all genes
involved downstream in the HER-2 signal transduction pathway
and apoptotic cell death. Furthermore, the combination of this
systemically administered, tumor-specific siRNA nanocomplex and
gemcitabine was able to inhibit significantly tumor growth of
established PANC-1 xenograft tumors.
Although the field of RNAi has made the transition from basic
research to clinical application for localized disease such as
macular degeneration in less than 10 years, a time frame perhaps
faster than that of any other approach in gene medicine, the lack of
efficient targeted delivery, low transfection efficiency, instability to
nucleases, poor tissue penetration, and nonspecific immune
stimulation have hindered siRNA from reaching its full therapeutic
Cancer Research
Cancer Res 2008; 68: (5). March 1, 2008
1248
www.aacrjournals.org
Research.
on June 11, 2013. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from
potential. This is particularly true for diseases such as cancer
where systemic delivery of targeted therapeutics is essential. In
addition to identifying the correct target gene and pathway,
effective anticancer siRNA therapies must also be able to deliver a
sufficient amount of intact, functional siRNA to the target cell.
Progress is being made to address these challenges, with several
approaches for cell-specific delivery for cancer, and to increase
transfection efficiency being reported. However, the vast majority
are still only applicable in vitro or for nonsystemic administration,
including intratumoral, i.m., and i.p. injection. The delivery system
described by Pirollo et al. (17) seems to be able to overcome the
limitations of the current technology. The complex itself is of
nanoparticle size and thus able to penetrate through the small
capillaries resulting in deeper tumor penetration. The siRNA is
a modified hybrid construct that may reduce off-target effects,
whereas the encapsulation of this siRNA within the liposome can
protect it from degradation and rapid renal clearance while in the
bloodstream. This approach has shown exquisite tumor-targeting
capabilities to primary and metastatic tumors. The TfR as a target
for tumor-specific delivery has been well documented in the
literature (20). The inclusion of the anti-TfRscFv also serves to
enhance transfection efficiency, as the receptor-bound complex is
internalized via receptor-mediated endocytosis. Once internalized,
the inclusion of the pH-sensitive, endosomal-disrupting peptide in
this nanocomplex enhances release of the payload, increasing the
effective cytoplasmic concentration of the siRNA, leading not only
to the efficient knockdown of the target as observed in the tumors
but, when used in combination with standard chemotherapeutic
agents, also to tumor growth inhibition. For maximum efficacy,
the use of this targeted siRNA delivery is envisioned not as a single
agent but as part of such a combinatorial treatment regimen.
Thus, combining all of these factors in one delivery vehicle may be
the means to advance the field beyond the current challenges.
Successful translation of this approach through clinical trials is
the next logical step toward the realization of the potential of
siRNA as anticancer therapeutics.
Acknowledgments
Received 10/9/2007; revised 10/31/2007; accepted 11/4/2007.
Grant support: SynerGene Therapeutics (K.F. Pirollo), National Foundation for
Cancer Research (E.H. Chang), and TriLink Research Award in the form of research
grade siRNAs (E.H. Chang).
We apologize to our colleagues whose outstanding publications have not been
directly cited due to space constraints.
Figure 1. In vivo tumor-specific fluorescence targeting in a metastatic mouse model. MDA435/LCC6 lung metastases were induced in female athymic nude mice by
the i.v. inoculation of 8
10
6
MDA435/LCC6 cells through the tail vein of female nude mice. Eight weeks after injection, the scL-HoKC/siRNA complex carrying
9 mg/kg of modified hybrid 6-FAM siRNA was i.v. injected into the mice. Three hours after i.v. tail vein injection, the animals were sacrificed, and tumor and other
organs were excised, photographed, and examined for fluorescence using a Nikon epifluorescence stereoscope. The identical field is shown in bright field and
fluorescence views with the arrow indicating metastases. Taken from Fig. 3B in Pirollo et al. (17).
Targeted Delivery of siRNA
www.aacrjournals.org
1249
Cancer Res 2008; 68: (5). March 1, 2008
Research.
on June 11, 2013. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from
References
1. Uprichard SL. The therapeutic potential of RNA
interference. FEBS Lett 2005;579:5996–6007.
2. Dallas A, Vlassov AV. RNAi: a novel antisense
technology and its therapeutic potential. Med Sci Monit
2006;12:RA67–74.
3. Meyer M, Wagner E. Recent developments in the
application of plasmid DNA-based vectors and small
interfering RNA therapeutics for cancer. Hum Gene
Ther 2006;17:1062–76.
4. Guo P. RNA nanotechnology: engineering, assembly
and applications in detection, gene delivery and therapy.
J Nanosci Nanotechnol 2005;5:1964–82.
5. Aagaard L, Rossi JJ. RNAi therapeutics: principles,
prospects and challenges. Adv Drug Deliv Rev 2007;59:
75–86.
6. Dykxhoorn DM, Lieberman J. The silent revolution:
RNA interference as basic biology, research tool, and
therapeutic. Annu Rev Med 2005;56:401–23.
7. Xie FY, Woodle MC, Lu PY. Harnessing in vivo siRNA
delivery for drug discovery and therapeutic develop-
ment. Drug Discov Today 2006;11:67–73.
8. De Paula D, Bentley MV, Mahato RI. Hydrophobization
and bioconjugation for enhanced siRNA delivery and
targeting. RNA 2007;13:431–56.
9. Schiffelers RM, Ansari A, Xu J, et al. Cancer siRNA
therapy by tumor selective delivery with ligand-targeted
sterically stabilized nanoparticle. Nucleic Acids Res
2004;32:e149.
10. Kim B, Tang Q, Biswas PS, et al. Inhibition of ocular
angiogenesis by siRNA targeting vascular endothelial
growth factor pathway genes: therapeutic strategy for
herpetic stromal keratitis. Am J Pathol 2004;165:2177–85.
11. Hu-Lieskovan S, Heidel JD, Bartlett DW, Davis ME,
Triche TJ. Sequence-specific knockdown of EWS-FLI1 by
targeted, nonviral delivery of small interfering RNA
inhibits tumor growth in a murine model of metastatic
Ewing’s sarcoma. Cancer Res 2005;65:8984–92.
12. Bartlett DW, Su H, Hildebrant IJ, Weber WA, Davis
ME. Impact of tumor-specific targeting on the
biodistribution and efficacy of siRNA nanoparticles
measured by multimodality in vivo imaging. PNAS
2007;104:15549–15.
13. Bartlett DW, Davis ME. Insights into the kinetics of
siRNA-mediated gene silencing from live-cell and live-
animal bioluminescent imaging. Nucleic Acids Res 2006;
34:322–33.
14. Song E, Zhu P, Lee SK, et al. Antibody mediated
in vivo delivery of small interfering RNAs via cell-surface
receptors. Nat Biotechnol 2005;23:709–17.
15. Hogrefe RI, Lebedev AV, Zon G, et al. Chemically
modified short interfering hybrids (siHYBRIDS): nano-
immunoliposome delivery in vitro and in vivo for RNAi
of HER-2. Nucleosides Nucleotides Nucleic Acids 2006;
25:889–907.
16. Pirollo KF, Zon G, Rait A, et al. Tumor-targeting
nanoimmunoliposome complex for short interfering
RNA delivery. Hum Gene Ther 2006;17:117–24.
17. Pirollo KF, Rait A, Zhou Q, et al. Materializing the
potential of small interfering RNA via a tumor-targeting
nanodelivery system. Cancer Res 2007;67:2938–43.
18. Haynes BF, Hemler M, Cotner T, et al. Characteriza-
tion of a monoclonal antibody (5E9) that defines a
human cell surface antigen of cell activation. J Immunol
1981;127:347–351.
19. Batra JK, Fitzgerald DJ, Chaudhary VK, Pastan I.
Single-chain immunotoxins directed at the human
transferrin receptor containing Pseudomonas exotoxin
A or diphtheria toxin: anti-TFR(Fv)-PE40 and DT388-
anti-TFR(Fv). Mol Cell Biol 1991;11:2200–5.
20. Daniels TR, Delgado T, Rodriguez JA, Helguera G,
Penichet ML. The transferrin receptor part I: biology
and targeting with cytotoxic antibodies for the treat-
ment of cancer. Clin Immunol 2006;121:144–58.
21. Xu L, Pirollo KF, Tang W, Rait A, Chang EH.
Transferrin-liposome-mediated systemic p53 gene ther-
apy in combination with radiation results in regression
of human head and neck cancer xenografts. Hum Gene
Ther 1999;10:2941–52.
22. Hohjoh H. RNA interference (RNA(i)) induction with
various types of synthetic oligonucleotide duplexes in
cultured human cells. FEBS Lett 2002;521:195–9.
23. Lamberton JS, Christian AT. Varying the nucleic acid
composition of siRNA molecules dramatically varies the
duration and degree of gene silencing. Mol Biotechnol
2003;24:111–20.
24. Judge AD, Sood V, Shaw JR, Fang D, McClintock K,
MacLachlan I. Sequence-dependent stimulation of the
mammalian innate immune response by synthetic
siRNA. Nat Biotechnol 2005;23:457–62.
25. Karanth H, Murthy RS. pH-sensitive liposomes—
principle and application in cancer therapy. J Pharm
Pharmacol 2007;59:469–83.
26. Chen QR, Zhang L, Luther PW, Mixson AJ. Optimal
transfection with the HK polymer depends on its degree
of branching and the pH of endocytic vesicles. Nucleic
Acids Res 2002;30:1338–45.
27. Aoki Y, Hosaka S, Kawa S, Kiyosawa K. Potential
tumor-targeting peptide vector of histidylated oligoly-
sine conjugated to a tumor-homing RGD motif. Cancer
Gene Ther 2001;8:783–7.
Cancer Research
Cancer Res 2008; 68: (5). March 1, 2008
1250
www.aacrjournals.org
Research.
on June 11, 2013. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from
... The anti-HER-2 siRNA was also successfully internalized into the SKBR-3 cells, which overexpresses the HER-2, with low levels (<25%) of HER-2 gene silencing [60]. Liposomes can be designed such that they are temperature sensitive and will react to external stimuli which will result in the disruption of the liposome and hence, deliver the encapsulated drug or gene [61]. Another strategy to enhance the therapeutic index of liposomes includes the addition of targeting ligands, which will interact with cell surface receptors that are upregulated in BC cells. ...
... This will improve cellular internalization by receptor-mediated endocytosis. For example, the results of a study involving the formulation of anti-HER2 immunoliposomes with a recombinant humanized anti-HER2 monoclonal antibody, anti-transferrin receptor antibody and siRNA have shown that the immunoliposomes were successfully taken up by BC animal models, and were more effective against these BC cells that overexpressed HER2 as compared with the nontargeted liposomes [61][62][63]. ...
Article
Globally, breast cancer is the second leading cause of cancer-related mortality among women, with approximately 1.4 million new cases diagnosed annually. Associated genetic perturbations are emerging in the face of intense scientific enquiry, facilitating its classification, prognostication and treatment. RNAi, utilizing siRNA, is a powerful treatment strategy to silence disease-causing genes. However, therapeutic siRNA instability and poor cellular uptake have limited its clinical application, necessitating the use of nanocarriers. In this review, we highlight the RNAi mechanism, HER-2/neu and MYC as breast cancer gene targets, and nonviral nanocarriers as potentially safe and efficient delivery systems.
... At present, the most common method of delivery is the use of lipid-based nanocarriers in which RNAi agents, such as siRNA and miRNA, are packaged. This method allows the agent to cross the cell membrane [57,58]. In future research, miRNA may be blocked using siRNA to trigger the suppression of the synthesis of many proteins [56]. ...
Article
Full-text available
Atherosclerosis is one of the leading causes of mortality from cardiovascular disease (CVD) and is a chronic inflammatory disease of the middle and large arteries caused by a disruption of lipid metabolism. Noncoding RNA (ncRNA), including microRNA (miRNA), small interfering RNA (siRNA) and long noncoding RNA (lncRNA), was investigated for the treatment of atherosclerosis. Regulation of the expression of noncoding RNA targets the constituent element of the pathogenesis of atherosclerosis. Currently, miRNA therapy commonly employs miRNA antagonists and mimic compounds. In this review, attention is focused on approaches to correcting molecular disorders based on the genetic regulation of the transcription of key genes responsible for the development of atherosclerosis. Promising technologies were considered for the treatment of atherosclerosis, and examples are given for technologies that have been shown to be effective in clinical trials.
... The scL nanocomplexes are capable of encapsulating diverse therapeutic payloads ranging from plasmid DNAs for gene therapy [30,87,108,109] to antisense oligonucleotides, siRNAs, miRNAs to modulate cellular gene expression [111][112][113][114] to small molecule therapeutic agents like temozolomide (TMZ) [115] or other small chemotherapeutics [116]. Virtually all cancer cells examined display increased expression of TfRs compared to normal cells and the TfR pathway has long been recognized as promising for targeting therapeutics to tumors [117,118]. ...
... To examine the therapeutic potential of miR-30a-5p in HNSCC in vivo, we formulated a miR-30a-5p mimic with novel modifications into a nanodelivery system (scL) bearing an antibody fragment (TfRscFv) that targets transferrin receptor on tumor cells (26,27). We confirmed that the scL carrier containing FITC-conjugated control oligonucleotide undergoes preferential uptake in HNSCC xenografts, when com-pared with lung or liver (Fig. 5A). ...
Article
Full-text available
Purpose: To identify deregulated and inhibitory miRNAs and generate novel mimics for replacement nanomedicine for head and neck squamous cell carcinomas (HNSCC). Experimental design: We integrated miRNA and mRNA expression, copy number variation, and DNA methylation results from The Cancer Genome Atlas (TCGA), with a functional genome-wide screen. Results: We reveal that the miR-30 family is commonly repressed, and all 5 members sharing these seed sequence similarly inhibit HNSCC proliferation in vitro. We uncover a previously unrecognized inverse relationship with overexpression of a network of important predicted target mRNAs deregulated in HNSCC, that includes key molecules involved in proliferation (EGFR, MET, IGF1R, IRS1, E2F7), differentiation (WNT7B, FZD2), adhesion, and invasion (ITGA6, SERPINE1). Reexpression of the most differentially repressed family member, miR-30a-5p, suppressed this mRNA program, selected signaling proteins and pathways, and inhibited cell proliferation, migration, and invasion in vitro. Furthermore, a novel miR-30a-5p mimic formulated into a targeted nanomedicine significantly inhibited HNSCC xenograft tumor growth and target growth receptors EGFR and MET in vivo. Significantly decreased miR-30a/e family expression was related to DNA promoter hypermethylation and/or copy loss in TCGA data, and clinically with decreased disease-specific survival in a validation dataset. Strikingly, decreased miR-30e-5p distinguished oropharyngeal HNSCC with poor prognosis in TCGA (P = 0.002) and validation (P = 0.007) datasets, identifying a novel candidate biomarker and target for this HNSCC subset. Conclusions: We identify the miR-30 family as an important regulator of signal networks and tumor suppressor in a subset of HNSCC patients, which may benefit from miRNA replacement nanomedicine therapy.
Article
Rheumatoid arthritis (RA) is a chronic, prevalent, immune-mediated, inflammatory, joint disorder affecting millions of people worldwide. Despite current treatment options, many patients remain unable to achieve remission and suffer from comorbidities. Because of several comorbidities as well as its chronic nature, it diminishes the quality of patients' life and intensifies socioeconomic cargo. Consolidating peptides with immensely effective drug delivery systems has the ability to alleviate adverse effects associated with conventional treatments. Peptides are widely used as targeting moieties for the delivery of nanotherapeutics. The use of novel peptide-based nanotherapeutics may open up new avenues for improving efficacy by promoting drug accumulation in inflamed joints and reducing off-target cytotoxicity. Peptide therapeutics have grabbed significant attention due to their advantages over small drug molecules as well as complex targeting moieties. In light of this, the market for peptide-based medications is growing exponentially. Peptides can provide the versatility required for the successful delivery of drugs due to their structural diversity and their capability to lead drugs at the site of inflammation while maintaining optimum therapeutic efficacy. This comprehensive review aims to provide an enhanced understanding of recent advancements in the arena of peptide-based nanotherapeutics to strengthen targeted delivery for the effective management of rheumatoid arthritis. Additionally, various peptides having therapeutic roles in rheumatoid arthritis are summarized along with regulatory considerations for peptides.
Article
Full-text available
In order to obtain antineoplastic compounds and innovative formulations, new technologies and testing methods are continuously being developed. Unfortunately, besides cancer cells, chemotherapy also affects normal cells. An option to avoid toxicity is represented by the targeted cancer treatment using novel pharmaceutical dosage forms. Liposomes represent a relatively new pharmaceutical dosage form, used for their many advantages. In this article, the methods of liposomal preparation are mentioned, along with the classification and the latest improvements involving this pharmaceutical form. The bioavailability of conventional liposomes is currently improved by developing photodynamic liposomes, pH or temperature sensitive liposomes and targeted liposomes.
Article
The effective translocation of small interfering RNA (siRNA) across cell membranes has become one of the main challenges in gene silencing therapy. In this study, we have carried out molecular dynamics simulations to investigate a systematic procedure with different carriers that could be convenient for efficient siRNA delivery into the cell. Starting with poly-amido-amine (PAMAM) dendrimers and cholesterol molecules as carriers, we have found cholesterol as the most efficient carrier for siRNA when it is covalently attached with the siRNA terminal group. Our simulations show that binding of this complex in the lipid membrane alters the structure and dynamics of the nearby lipids to initiate the translocation process. Potential of mean force (PMF) was computed for siRNA with the carriers along the bilayer normal to understand the spontaneity of the process. Though all the PMF profiles show repulsive interaction inside the bilayer, the siRNA with cholesterol shows a comparative attractive interaction (∼27 kcal/mol) with respect to the siRNA-PAMAM complex. Altogether, our results demonstrate the binding interaction of the siRNA-carrier complex in the lipid membrane and propose a theoretical model for the efficient carrier by comparative study of the binding. The probable mechanism of the translocation process is also provided by the alteration of the lipid structure and dynamics for specifically siRNA-cholesterol binding.
Article
Full-text available
ماکرومولکول هاي زيستي اعم از RNA، DNA و پروتئين ها به دليل دارا بودن پتانسيل هاي ذاتي ويژه، به عنوان واحدهاي ساختاري براي تهيه نانوابزارها، بکار گرفته مي شوند. پروتئين ها در نانوزيست فناوري بصورت گسترده مورد استفاده قرار گرفته اند و نانوفناوري DNA نيز از جوانب بسياري مطالعه شده است. اما مفهوم نانوفناوري RNA اخيرا مطرح گرديده است و اولين شواهد مبني بر ساخت نانوذرات RNA از طريق فرايند خودساماندهي چندين مولکول RNA طبيعي يا مهندسي شده، در سال 1998 مطرح شد. از آنجايي که مولکول هاي RNA به مانند DNA به سهولت قابل دست ورزي بوده و مثل پروتئين ها داراي تنوع بسيار در ساختار و عملکرد خود هستند، در نانوزيست فناوري بسيار مورد توجه واقع شده اند. نانوذرات RNA طي فرايند خودسامانده و با استفاده از ميانکنش هاي هماهنگ، بين مولکول هاي RNA گرد هم مي آيند و ساختارهاي دو يا سه بعدي را ايجاد مي کنند. اولين و بهترين RNA با کاربرد نانوفناوري که تاکنون شناخته شده، موتور packing RNA باکتريوفاژ j29 است که قابليت ايجاد ديمر، تريمر و هگزامر را داراست. به اين نانوذرات، مي توان مولکول هاي درماني، تشخيصي و گيرنده ها، اعم از siRNA ها، ريبوزيم ها و آپتامرهاي RNA را مت‍صل نمود. از جمله عوامل درماني قابل استفاده، siRNAهاي طراحي شده عليه سرطان ريه است. در اين سرطان، بافت بدخيم در يک يا هر دو ريه رشد مي کند. سرطان ريه يکي از همه گيرترين سرطان ها در سراسر جهان است و بيش از 80% بيماران در کمتر از 5 سال از زمان شناسايي بيماري، جان خود را از دست مي دهند. از بين همه siRNAهاي مورد استفاده براي درمان سرطان ريه، siRNAهاي ژن هاي KRAS و P53 تاثير بيشتري در بهبودي اين بيماري نشان داده اند. در اين مقاله، مفاهيم مربوط به ساختار و عملکرد RNAهاي مورد استفاده در نانوزيست فناوري، رويکردهاي مورد استفاده به منظور مهندسي يا ساخت اين RNAها و تبديل آنها به نانوذرات کاربردي مرور گشته و نقش آنها در درمان سرطان ريه بررسي شده است.‬
Article
Full-text available
Antisense oligonucleotide agents induce the inhibition of target gene expression in a sequence-spe-cifi c manner by exploiting the ability of oligonucleotides to bind to target RNAs via Watson-Crick hybridization. Once bound, the antisense agent either disables or induces the degradation of the target RNA. This technology may be used for therapeutic purposes, functional genomics, and target validation. There are three major categories of gene-silencing molecules: (1) antisense oligo-nucleotide derivatives that, depending on their type, recruit RNase H to cleave the target mRNA or inhibit translation by steric hindrance; (2) ribozymes and deoxyribozymes-catalytically active oligonucleotides that cause RNA cleavage; (3) small interfering double-stranded RNA molecules that induce RNA degradation through a natural gene-silencing pathway called RNA interference (RNAi). RNAi is the latest addition to the family of antisense technologies and has rapidly become the most widely used approach for gene knockdown because of its potency. In this mini-review, we introduce the RNAi effect, briefl y compare it with existing antisense technologies, and discuss its therapeutic potential, focusing on recent animal studies and ongoing clinical trials. RNAi may provide new therapeutics for treating viral infections, neurodegenerative diseases, septic shock, mac-ular degeneration, cancer, and other illnesses, although in vivo delivery of small interfering RNAs remains a signifi cant obstacle. key words: RNA interference • siRNA • antisense technology • animal studies Full-text PDF:
Article
Full-text available
The use of cationic liposomes as nonviral vehicles for the delivery of therapeutic molecules is becoming increasingly prevalent in the field of gene therapy. We have previously demonstrated that the use of the transferrin ligand (Tf) to target a cationic liposome delivery system resulted in a significant increase in the transfection efficiency of the complex [Xu, L., Pirollo, K.F., and Chang, E.H. (1997). Hum. Gene Ther. 8, 467-475]. Delivery of wild-type (wt) p53 to a radiation-resistant squamous cell carcinoma of the head and neck (SCCHN) cell line via this ligand-targeted, liposome complex was also able to revert the radiation resistant phenotype of these cells in vitro. Here we optimized the Tf/liposome/DNA ratio of the complex (LipT) for maximum tumor cell targeting, even in the presence of serum. The efficient reestablishment of wtp53 function in these SCCHN tumor cells in vitro, via the LipT complex, restored the apoptotic pathway, resulting in a significant increase in radiation-induced apoptosis that was directly proportional to the level of exogenous wtp53 in the tumor cells. More significantly, intravenous administration of LipT-p53 markedly sensitized established SCCHN nude mouse xenograft tumors to radiotherapy. The combination of systemic LipT-p53 gene therapy and radiation resulted in complete tumor regression and inhibition of their recurrence even 6 months after the end of all treatment. These results indicate that this tumor-specific, ligand-liposome delivery system for p53 gene therapy, when used in concert with conventional radiotherapy, can provide a new and more effective means of cancer treatment.
Article
Full-text available
Two single-chain immunotoxins directed at the human transferrin receptor have been constructed by using polymerase chain reaction-based methods. Anti-TFR(Fv)-PE40 is encoded by a gene fusion between the DNA sequence encoding the antigen-binding portion (Fv) of a monoclonal antibody directed at the human transferrin receptor and that encoding a 40,000-molecular-weight fragment of Pseudomonas exotoxin (PE40). The other fusion protein, DT388-anti-TFR(Fv), is encoded by a gene fusion between the DNA encoding a truncated form of diphtheria toxin and that encoding the antigen-binding portion of antibody to human transferrin receptor. These gene fusions were expressed in Escherichia coli, and fusion proteins were purified by conventional chromatography techniques to near homogeneity. In anti-TFR(Fv)-PE40, the antigen-binding portion is placed at the amino terminus of the toxin, while in DT388-anti-TFR(Fv), it is at the carboxyl end of the toxin. Both these single-chain immunotoxins kill cells bearing the human transferrin receptors. However, anti-TFR(Fv)-PE40 was usually more active than DT388-anti-TFR(Fv), and in some cases it was several-hundred-fold more active. Anti-TFR(Fv)-PE40 was also more active on cell lines than a conjugate made by chemically coupling the native antibody to PE40, and in some cases it was more than 100-fold more active.
Article
Full-text available
We have recently reported that liposomes in combination with histidine (HK)-containing polymers enhanced the expression of luciferase in transfected cells. In transformed or malignant cell lines, branched HK polymers (combined with liposome carriers) were significantly more effective than the linear HK polymer in stimulating gene expression. In the current study, we found that the linear HK polymer enhanced gene expression in primary cell lines more effectively than the branched polymers. The differences in the optimal carrier (linear versus branched) were not due to initial cellular uptake, size of the complexes or level of gene expression. There was, however, a strong association between the optimal type of HK polymer and the pH of endocytic vesicles (P = 0.0058). By altering the percentage of histidines carrying a positive charge, the endosomal pH of a cell may determine the amount of DNA released from the linear or branched HK polymer. In the two cell lines in which the linear HK was the optimal polymer, the endocytic vesicles were strongly acidic with a pH of <5.0. Conversely, in the four cell lines in which the branched polymers were optimal transfection agents, the pH of endocytic vesicles was >6.0. Furthermore, binding data support the relationship between DNA release from the optimal HK polymer and endosomal pH. The interplay between optimal HK polymers and the endosomal pH may lead to improved gene-delivery polymers tailored to a particular cell.
Article
We have recently reported that liposomes in combination with histidine (HK)-containing polymers enhanced the expression of luciferase in transfected cells. In transformed or malignant cell lines, branched HK polymers (combined with liposome carriers) were significantly more effective than the linear HK polymer in stimulating gene expression. In the current study, we found that the linear HK polymer enhanced gene expression in primary cell lines more effectively than the branched polymers. The differences in the optimal carrier (linear versus branched) were not due to initial cellular uptake, size of the complexes or level of gene expression. There was, however, a strong association between the optimal type of HK polymer and the pH of endocytic vesicles (P = 0.0058). By altering the percentage of histidines carrying a positive charge, the endosomal pH of a cell may determine the amount of DNA released from the linear or branched HK polymer. In the two cell lines in which the linear HK was the optimal polymer, the endocytic vesicles were strongly acidic with a pH of <5.0. Conversely, in the four cell lines in which the branched polymers were optimal transfection agents, the pH of endocytic vesicles was >6.0. Furthermore, binding data support the relationship between DNA release from the optimal HK polymer and endosomal pH. The interplay between optimal HK polymers and the endosomal pH may lead to improved gene-delivery polymers tailored to a particular cell.
Article
This study describes the monoclonal antibody 5E9 and the cell surface antigen it defines. The hybridoma cell line T3-5E9 was derived from fusion of P3 X 63/Ag8 myeloma cells and spleen cells from BALB/c mice immunized with HSB-2 cells, a human T cell line. Although binding to only 1 to 5% of peripheral blood (PB) and spleen mononuclear cells, 5E9 antibody bound to 40 to 80% of Con A-, PHA-, or PWM-activated PB cells. Moreover, 5E9 antibody bound to variable numbers of Sezary, acute myelogenous leukemia, and ALL PB leukemia cells. 5E9 antibody bound to all hematopoietic and nonhematopoietic cell lines tested, to 11 +/- 1% of thymocytes, and 40% of nucleated bone marrow cells. Under reducing conditions, immunoprecipitation studies using 5E9 antibody demonstrated 5E9 antigen to be an 90,000 m.w. glycoprotein. Under nonreducing conditions, antigen 5E9 is a disulfide-linked dimer of approximately 190,000 daltons. Sequential precipitation experiments using antibody 5E9, alpha OD heteroantiserum (raised against T ALL cells), and monoclonal antibody OKT9 demonstrated that the 3 antibody preparations recognized the same 90,000 m.w. glycoprotein. Thus, antibody 5E9 defines an 90,000 m.w. human cell surface antigen that is absent on the majority of PB mononuclear cells and is expressed on rapidly dividing normal and malignant human cells. This monoclonal antibody should be a useful marker of human cell activation.
Article
We have developed a potential tumor-targeting peptide vector (cRGD-hK) that is intended to be systemically and repeatedly administered to patients with advanced solid tumors. The peptide vector of 36 l-amino acid residues, CRGDCF(K[H-]KKK)6, comprises a tumor-homing RGD motif, a DNA-binding oligolysine, and histidyl residues to facilitate the delivery into the cytosol. Using cytomegalovirus-driven luciferase expression plasmids as a reporter, we tested the transfection efficiency of cRGD-hK in hepatoma and pancreatic cancer cell lines. Transfection with the cRGD-hK/plasmid complexes (molar ratio 4000:1) was inhibited by 50 nM bafilomycin A1, an inhibitor of the vacuolar ATPase endosomal proton pump, or 10 microM cycloRGDfV, an integrin alphavbeta3 antagonist, indicating that the three elements of cRGD-hK could function as expected, at least in vitro. In nude mice bearing tumors created by subcutaneous inoculation, luciferase activity in the tumor tissues 48 hours after the injection of the cRGD-hK/plasmid complexes through the tail vein (20 microg plasmids per mouse) was significantly higher than that in the lung, kidney, and spleen, but only slightly higher than that in the liver. Although the latter difference was small, we propose a potential nonviral gene therapy for advanced solid tumors through use of the tumor-targeting peptide vector.
Article
Various types of synthetic oligonucleotide duplexes against the Photinus luciferase gene were tested on their induction of the sequence-specific RNA interference (RNAi) activity in transfected human cells. Results indicate that RNA duplexes with ribonucleotide 3' overhangs rather than those with deoxyribonucleotide 3' overhangs induce more efficient RNAi activity, and that sense-stranded DNA/antisense-stranded RNA hybrids induce a moderate RNAi activity. These results suggest that there is a difference in the potential of oligonucleotide duplexes to be RNAi mediators, i.e. short interfering RNAs (siRNAs), between human RNAi and invertebrate RNAi. The data further show that different siRNAs induce different levels of RNAi.
Article
The utility of short interfering RNA (siRNA) as a means of gene silencing depends on several factors. These include the degree to which a gene can be silenced, the length of time for which the gene remains silenced, the degree of recovery of gene function, and the effects of the silencing process on general cell functions. We hypothesized that changing the nucleic acid composition of the siRNA constructs used for silencing would affect these parameters. With siRNA gene silencing of the glucose-6-phosphate dehydrogenase gene as a baseline, we found that siDNA molecules have an effect that is similar in duration but lesser in degree, whereas hybrid DNA:RNA molecules have an effect that is enormously greater in both duration and degree.