Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
EMBO Mol Med ; 11(12): e10923, 2019 12.
Article in English | MEDLINE | ID: mdl-31709774

ABSTRACT

High extracellular matrix (ECM) content in solid cancers impairs tumour perfusion and thus access of imaging and therapeutic agents. We have devised a new approach to degrade tumour ECM, which improves uptake of circulating compounds. We target the immune-modulating cytokine, tumour necrosis factor alpha (TNFα), to tumours using a newly discovered peptide ligand referred to as CSG. This peptide binds to laminin-nidogen complexes in the ECM of mouse and human carcinomas with little or no peptide detected in normal tissues, and it selectively delivers a recombinant TNFα-CSG fusion protein to tumour ECM in tumour-bearing mice. Intravenously injected TNFα-CSG triggered robust immune cell infiltration in mouse tumours, particularly in the ECM-rich zones. The immune cell influx was accompanied by extensive ECM degradation, reduction in tumour stiffness, dilation of tumour blood vessels, improved perfusion and greater intratumoral uptake of the contrast agents gadoteridol and iron oxide nanoparticles. Suppressed tumour growth and prolonged survival of tumour-bearing mice were observed. These effects were attainable without the usually severe toxic side effects of TNFα.


Subject(s)
Extracellular Matrix/metabolism , Animals , Cell Line , Cell Surface Display Techniques , Contrast Media/metabolism , Female , Ferric Compounds/metabolism , Gadolinium/metabolism , Heterocyclic Compounds/metabolism , Humans , Male , Mice , Nanoparticles/metabolism , Organometallic Compounds/metabolism , Tumor Necrosis Factor-alpha/metabolism
2.
Nat Biomed Eng ; 2(2): 95-103, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29955439

ABSTRACT

Bacterial resistance to antibiotics has made it necessary to resort to antibiotics that have considerable toxicities. Here, we show that the cyclic 9-amino acid peptide CARGGLKSC (CARG), identified via phage display on Staphylococcus aureus (S. aureus) bacteria and through in vivo screening in mice with S. aureus-induced lung infections, increases the antibacterial activity of CARG-conjugated vancomycin-loaded nanoparticles in S. aureus-infected tissues and reduces the needed overall systemic dose, minimizing side effects. CARG binds specifically to S. aureus bacteria but not Pseudomonas bacteria in vitro, selectively accumulates in S. aureus-infected lungs and skin of mice but not in non-infected tissue and Pseudomonas-infected tissue, and significantly enhances the accumulation of intravenously injected vancomycin-loaded porous silicon nanoparticles bearing the peptide in S. aureus-infected mouse lung tissue. The targeted nanoparticles more effectively suppress staphylococcal infections in vivo relative to equivalent doses of untargeted vancomycin nanoparticles or of free vancomycin. The therapeutic delivery of antibiotic-carrying nanoparticles bearing peptides targeting infected tissue may help combat difficult-to-treat infections.

3.
Nat Commun ; 9(1): 1070, 2018 03 09.
Article in English | MEDLINE | ID: mdl-29523838

ABSTRACT

The original version of the Supplementary Information associated with this Article inadvertently omitted Supplementary Table 1. The HTML has now been updated to include a corrected version of the Supplementary Information.

4.
Nat Commun ; 8(1): 1403, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29123083

ABSTRACT

Cerebrovascular changes occur in Alzheimer's disease (AD). Using in vivo phage display, we searched for molecular markers of the neurovascular unit, including endothelial cells and astrocytes, in mouse models of AD. We identified a cyclic peptide, CDAGRKQKC (DAG), that accumulates in the hippocampus of hAPP-J20 mice at different ages. Intravenously injected DAG peptide homes to neurovascular unit endothelial cells and to reactive astrocytes in mouse models of AD. We identified connective tissue growth factor (CTGF), a matricellular protein that is highly expressed in the brain of individuals with AD and in mouse models, as the target of the DAG peptide. We also showed that exogenously delivered DAG homes to the brain in mouse models of glioblastoma, traumatic brain injury, and Parkinson's disease. DAG may potentially be used as a tool to enhance delivery of therapeutics and imaging agents to sites of vascular changes and astrogliosis in diseases associated with neuroinflammation.


Subject(s)
Alzheimer Disease/metabolism , Peptides, Cyclic/metabolism , Alzheimer Disease/etiology , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Astrocytes/metabolism , Astrocytes/pathology , Brain/blood supply , Brain/metabolism , Brain/pathology , Connective Tissue Growth Factor/metabolism , Disease Models, Animal , Hippocampus/blood supply , Hippocampus/metabolism , Hippocampus/pathology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Peptide Library , Peptides, Cyclic/chemistry , Protein Binding
5.
J Control Release ; 268: 49-56, 2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29030222

ABSTRACT

Anti-angiogenic and vascular disrupting therapies rely on the dependence of tumors on new blood vessels to sustain tumor growth. We previously reported a potent vascular disrupting agent, a theranostic nanosystem consisting of a tumor vasculature-homing peptide (CGKRK) fused to a pro-apoptotic peptide [D(KLAKLAK)2] coated on iron oxide nanoparticles. This nanosystem showed promising therapeutic efficacy in glioblastoma (GBM) and breast cancer models. However, complete control of the tumors was not achieved, and some tumors became non-responsive to the treatment. Here we examined the non-responder phenomenon in an aggressive MCF10-CA1a breast tumor model. In the treatment-resistant tumors we noted the emergence of CD31-negative patent neovessels and a concomitant loss of tumor homing of the nanosystem. In vivo phage library screening in mice bearing non-responder tumors showed that compared to untreated and treatment-sensitive tumors, treatment sensitive tumors yield a distinct landscape of vascular homing peptides characterized by over-representation of peptides that target αv integrins. Our approach may be generally applicable to the development of targeted therapies for tumors that have failed treatment.


Subject(s)
Breast Neoplasms/drug therapy , Nanoparticles/therapeutic use , Neovascularization, Pathologic/drug therapy , Peptides/therapeutic use , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/pathology , Peptide Library
6.
Elife ; 62017 03 23.
Article in English | MEDLINE | ID: mdl-28332979

ABSTRACT

Regeneration of skeletal muscle in adults is mediated by satellite stem cells. Accumulation of misfolded proteins triggers endoplasmic reticulum stress that leads to unfolded protein response (UPR). The UPR is relayed to the cell through the activation of PERK, IRE1/XBP1, and ATF6. Here, we demonstrate that levels of PERK and IRE1 are increased in satellite cells upon muscle injury. Inhibition of PERK, but not the IRE1 arm of the UPR in satellite cells inhibits myofiber regeneration in adult mice. PERK is essential for the survival and differentiation of activated satellite cells into the myogenic lineage. Deletion of PERK causes hyper-activation of p38 MAPK during myogenesis. Blocking p38 MAPK activity improves the survival and differentiation of PERK-deficient satellite cells in vitro and muscle formation in vivo. Collectively, our results suggest that the PERK arm of the UPR plays a pivotal role in the regulation of satellite cell homeostasis during regenerative myogenesis.


Subject(s)
Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Regeneration , Satellite Cells, Skeletal Muscle/physiology , Unfolded Protein Response , eIF-2 Kinase/metabolism , Animals , Membrane Proteins/metabolism , Mice , Muscle, Skeletal/injuries , Protein Serine-Threonine Kinases/metabolism
7.
Bio Protoc ; 6(19)2016 Oct 05.
Article in English | MEDLINE | ID: mdl-27819014

ABSTRACT

Adult skeletal muscle regeneration is orchestrated by a specialized population of adult stem cells called satellite cells, which are localized between the basal lamina and the plasma membrane of myofibers. The process of satellite cell-activation, proliferation, and subsequent differentiation that occurs during muscle regeneration can be recapitulated ex vivo by isolation of single myofibers from skeletal muscles and culturing them under suspension conditions. Here, we describe an improved protocol to evaluate ex vivo satellite cells activation through isolation of single myofibers from extensor digitorum longus (EDL) muscle of mice and culturing and staining of myofiber-associated satellite cells with the markers of self-renewal, proliferation, and differentiation.

8.
Nat Commun ; 7: 11980, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27351915

ABSTRACT

Traumatic brain injury (TBI) is a major health and socio-economic problem, but no pharmacological agent is currently approved for the treatment of acute TBI. Thus, there is a great need for advances in this field. Here, we describe a short peptide (sequence CAQK) identified by in vivo phage display screening in mice with acute brain injury. The CAQK peptide selectively binds to injured mouse and human brain, and systemically injected CAQK specifically homes to sites of brain injury in mouse models. The CAQK target is a proteoglycan complex upregulated in brain injuries. Coupling to CAQK increased injury site accumulation of systemically administered molecules ranging from a drug-sized molecule to nanoparticles. CAQK-coated nanoparticles containing silencing oligonucleotides provided the first evidence of gene silencing in injured brain parenchyma by systemically administered siRNA. These findings present an effective targeting strategy for the delivery of therapeutics in clinical management of acute brain injuries.


Subject(s)
Brain Injuries, Traumatic/therapy , Drug Delivery Systems , Peptides , Aged , Animals , Brain Injuries, Traumatic/diagnostic imaging , Extracellular Matrix/metabolism , Humans , Male , Mice , Middle Aged
9.
Nanoscale Horiz ; 1(5): 407-414, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-29732165

ABSTRACT

We report the synthesis, characterization, and assessment of a nanoparticle-based RNAi delivery platform that protects siRNA payloads against nuclease-induced degradation and efficiently delivers them to target cells. The nanocarrier is based on biodegradable mesoporous silicon nanoparticles (pSiNPs), where the voids of the nanoparticles are loaded with siRNA and the nanoparticles are encapsulated with graphene oxide nanosheets (GO-pSiNPs). The graphene oxide encapsulant delays release of the oligonucleotide payloads in vitro by a factor of 3. When conjugated to a targeting peptide derived from the rabies virus glycoprotein (RVG), the nanoparticles show 2-fold greater cellular uptake and gene silencing. Intravenous administration of the nanoparticles into brain-injured mice results in substantial accumulation specifically at the site of injury.

10.
Toxicol Appl Pharmacol ; 287(1): 86-92, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26048585

ABSTRACT

The medical applications of aptamers have recently emerged. We developed an antagonistic thioaptamer (ESTA) against E-selectin. Previously, we showed that a single injection of ESTA at a dose of 100µg inhibits breast cancer metastasis in mice through the functional blockade of E-selectin. In the present study, we evaluated the safety of different doses of intravenously administered ESTA in single-dose acute and repeat-dose subacute studies in ICR mice. Our data indicated that intravenous administration of up to 500µg ESTA did not result in hematologic abnormality in either study. Additionally, intravenous injection of ESTA did not affect the levels of plasma cytokines (IL-1ß, IL-2, IL-4, IL-5, IL-6, IL-10, GM-CSF, IFN-γ, and TNF-α) or complement split products (C3a and C5a) in either study. However, repeated injections of ESTA slightly increased plasma ALT and AST activities, in accordance with the appearance of small necrotic areas in the liver. In conclusion, our data demonstrated that intravenous administration of ESTA does not cause overt hematologic, organs, and immunologic responses under the experimental conditions.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/administration & dosage , E-Selectin/drug effects , Alanine Transaminase/blood , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/toxicity , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/toxicity , Aspartate Aminotransferases/blood , Biomarkers/blood , Chemical and Drug Induced Liver Injury/blood , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Complement C3a/metabolism , Complement C5a/metabolism , Cytokines/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , E-Selectin/metabolism , Female , Injections, Intravenous , Kidney/drug effects , Liver/drug effects , Liver/pathology , Male , Mice, Inbred ICR , Necrosis , Risk Assessment
11.
Mol Ther ; 23(6): 1044-1054, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25815697

ABSTRACT

Shear-resistant adhesion and extravasation of disseminated cancer cells at the target organ is a crucial step in hematogenous metastasis. We found that the vascular adhesion molecule E-selectin preferentially promoted the shear-resistant adhesion and transendothelial migration of the estrogen receptor (ER)(-)/CD44(+) hormone-independent breast cancer cells, but not of the ER(+)/CD44(-/low) hormone-dependent breast cancer cells. Coincidentally, CD44(+) breast cancer cells were abundant in metastatic lung and brain lesions in ER(-) breast cancer, suggesting that E-selectin supports hematogenous metastasis of ER(-)/CD44(+) breast cancer. In an attempt to prevent hematogenous metastasis through the inhibition of a shear-resistant adhesion of CD44(+) cancer cells to E-selectin-expressing blood vessels on the premetastatic niche, an E-selectin targeted aptamer (ESTA) was developed. We demonstrated that a single intravenous injection of ESTA reduced metastases to a baseline level in both syngeneic and xenogeneic forced breast cancer metastasis models without relocating the site of metastasis. The effect of ESTA was absent in E-selectin knockout mice, suggesting that E-selectin is a molecular target of ESTA. Our data highlight the potential application of an E-selectin antagonist for the prevention of hematogenous metastasis of ER(-)/CD44(+) breast cancer.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis/prevention & control , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Cell Adhesion , Cell Line, Tumor , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/metabolism , Female , Genetic Therapy , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Neoplasm Metastasis/genetics , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Transendothelial and Transepithelial Migration/genetics
12.
Biomed Res Int ; 2014: 727459, 2014.
Article in English | MEDLINE | ID: mdl-25276814

ABSTRACT

Aged keratinocytes have diminished proliferative capacity and hyaluronan (HA) cell coats, which are losses that contribute to atrophic skin characterized by reduced barrier and repair functions. We formulated HA-phospholipid (phosphatidylethanolamine, HA-PE) polymers that form pericellular coats around cultured dermal fibroblasts independently of CD44 or RHAMM display. We investigated the ability of these HA-PE polymers to penetrate into aged mouse skin and restore epidermal function in vivo. Topically applied Alexa(647)-HA-PE penetrated into the epidermis and dermis, where it associated with both keratinocytes and fibroblasts. In contrast, Alexa(647)-HA was largely retained in the outer cornified layer of the epidermis and quantification of fluorescence confirmed that significantly more Alexa(647)-HA-PE penetrated into and was retained within the epidermis than Alexa(647)-HA. Multiple topical applications of HA-PE to shaved mouse skin significantly stimulated basal keratinocyte proliferation and epidermal thickness compared to HA or vehicle cream alone. HA-PE had no detectable effect on keratinocyte differentiation and did not promote local or systemic inflammation. These effects of HA-PE polymers are similar to those reported for endogenous epidermal HA in youthful skin and show that topical application of HA-PE polymers can restore some of the impaired functions of aged epidermis.


Subject(s)
Hyaluronic Acid/pharmacology , Keratinocytes/cytology , Keratinocytes/drug effects , Phosphatidylethanolamines/pharmacology , Polymers/pharmacology , Administration, Topical , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Epidermal Cells , Epidermis/drug effects , Epidermis/metabolism , Extracellular Matrix Proteins/deficiency , Extracellular Matrix Proteins/metabolism , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Hyaluronan Receptors/metabolism , Inflammation/pathology , Keratinocytes/metabolism , Mice, Inbred C57BL
13.
Nat Mater ; 13(9): 904-11, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24907927

ABSTRACT

There is considerable interest in using nanoparticles as labels or to deliver drugs and other bioactive compounds to cells in vitro and in vivo. Fluorescent imaging, commonly used to study internalization and subcellular localization of nanoparticles, does not allow unequivocal distinction between cell surface-bound and internalized particles, as there is no methodology to turn particles 'off'. We have developed a simple technique to rapidly remove silver nanoparticles outside living cells, leaving only the internalized pool for imaging or quantification. The silver nanoparticle (AgNP) etching is based on the sensitivity of Ag to a hexacyanoferrate-thiosulphate redox-based destain solution. In demonstration of the technique we present a class of multicoloured plasmonic nanoprobes comprising dye-labelled AgNPs that are exceptionally bright and photostable, carry peptides as model targeting ligands, can be etched rapidly and with minimal toxicity in mice, and that show tumour uptake in vivo.


Subject(s)
Cells/metabolism , Metal Nanoparticles , Molecular Imaging/methods , Molecular Probes/chemistry , Molecular Probes/metabolism , Silver/chemistry , Silver/metabolism , Animals , Avidin/chemistry , Biological Transport , Cell Line, Tumor , Female , Humans , Mice , Molecular Probes/analysis , Molecular Probes/toxicity , Polyethylene Glycols/chemistry , Silver/toxicity
14.
Methods Mol Biol ; 1049: 481-93, 2013.
Article in English | MEDLINE | ID: mdl-23913239

ABSTRACT

RNA interference (RNAi) is a powerful approach for silencing oncogenes; however, in vivo RNAi delivery has remained a major challenge due to lack of safe, efficient, and sustained delivery. Here, we describe a novel approach to overcome these limitations using mesoporous silicon particles loaded with nanoparticles (i.e., liposomes) containing small interfering RNA (siRNA) targeted against oncoprotein that contributes to cancer cell survival. This delivery method resulted in sustained gene silencing for at least 3 weeks with substantial reduction of tumor growth with no overt toxicities in two independent orthotopic mouse models of ovarian cancer following a single intravenous administration of mesoporous silicon particles loaded with liposomal EphA2-siRNA.


Subject(s)
Gene Silencing , Gene Transfer Techniques , Ovarian Neoplasms/genetics , RNA, Small Interfering/administration & dosage , Animals , Cell Line, Tumor , Female , Genetic Therapy/methods , Humans , Liposomes/administration & dosage , Mice , Nanoparticles/administration & dosage , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , RNA Interference , Silicon/administration & dosage
16.
Oncotarget ; 2(4): 298-304, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21666286

ABSTRACT

Recent developments in multi-functional nanoparticles offer a great potential for targeted delivery of therapeutic compounds and imaging contrast agents to specific cell types, in turn, enhancing therapeutic effect and minimizing side effects. Despite the promise, site specific delivery carriers have not been translated into clinical reality. In this study, we have developed long circulating liposomes with the outer surface decorated with thioated oligonucleotide aptamer (thioaptamer) against E-selectin (ESTA) and evaluated the targeting efficacy and PK parameters. In vitro targeting studies using Human Umbilical Cord Vein Endothelial Cell (HUVEC) demonstrated efficient and rapid uptake of the ESTA conjugated liposomes (ESTA-lip). In vivo, the intravenous administration of ESTA-lip resulted in their accumulation at the tumor vasculature of breast tumor xenografts without shortening the circulation half-life. The study presented here represents an exemplary use of thioaptamer and liposome and opens the door to testing various combinations of thioaptamer and nanocarriers that can be constructed to target multiple cancer types and tumor components for delivery of both therapeutics and imaging agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/therapeutic use , Drug Delivery Systems/methods , Liposomes/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/pharmacokinetics , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacokinetics , Breast Neoplasms/blood supply , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma/blood supply , Carcinoma/drug therapy , Carcinoma/pathology , Cells, Cultured , Drug Carriers/therapeutic use , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Humans , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Mice, Nude , Models, Biological , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/therapeutic use , Xenograft Model Antitumor Assays
17.
Cancer Sci ; 102(7): 1247-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21447010

ABSTRACT

Cancer is a leading cause of morbidity and mortality worldwide, with recent advancements resulting in modest impacts on patient survival. Nanomedicine represents an innovative field with immense potential for improving cancer treatment, having ushered in several established drug delivery platforms. Nanoconstructs such as liposomes are widely used in clinics, while polymer micelles are in advanced phases of clinical trials in several countries. Currently, the field of nanomedicine is generating a new wave of nanoscale drug delivery strategies, embracing trends that involve the functionalization of these constructs with moieties that enhance site-specific delivery and tailored release. Herein, we discuss several advancements in established nanoparticle technologies such as liposomes, polymer micelles, and dendrimers regarding tumor targeting and controlled release strategies, which are being incorporated into their design with the hope of generating a more robust and efficacious nanotherapeutic modality. We also highlight a novel strategy known as multistage drug delivery; a rationally designed nanocarrier aimed at overcoming numerous biological barriers involved in drug delivery through the decoupling of various tasks that comprise the journey from the moment of systemic administration to arrival at the tumor site.


Subject(s)
Antineoplastic Agents/administration & dosage , Nanomedicine , Neoplasms/drug therapy , Animals , Delayed-Action Preparations , Dendrimers , Drug Carriers , Drug Delivery Systems , Humans , Liposomes , Micelles , Nanoparticles
18.
PLoS One ; 5(9)2010 Sep 30.
Article in English | MEDLINE | ID: mdl-20927342

ABSTRACT

Active targeting of a drug carrier to a specific target site is crucial to provide a safe and efficient delivery of therapeutics and imaging contrast agents. E-selectin expression is induced on the endothelial cell surface of vessels in response to inflammatory stimuli but is absent in the normal vessels. Thus, E-selectin is an attractive molecular target, and high affinity ligands for E-selectin could be powerful tools for the delivery of therapeutics and/or imaging agents to inflamed vessels. In this study, we identified a thiophosphate modified aptamer (thioaptamer, TA) against E-selectin (ESTA-1) by employing a two-step selection strategy: a recombinant protein-based TA binding selection from a combinatorial library followed by a cell-based TA binding selection using E-selectin expressing human microvascular endothelial cells. ESTA-1 selectively bound to E-selectin with nanomolar binding affinity (K(D) = 47 nM) while exhibiting minimal cross reactivity to P- and L-selectin. Furthermore, ESTA-1 binding to E-selectin on the endothelial cells markedly antagonized the adhesion (over 75% inhibition) of sLe(x) positive HL-60 cells at nanomolar concentration. ESTA-1 also bound specifically to the inflamed tumor-associated vasculature of human carcinomas derived from breast, ovarian, and skin but not to normal organs, and this binding was highly associated with the E-selectin expression level. Similarly, intravenously injected ESTA-1 demonstrated distinct binding to the tumor vasculature in a breast cancer xenograft model. Together, our data substantiates the discovery of a thioaptamer (ESTA-1) that binds to E-selectin with high affinity and specificity, thereby highlighting the potential application of ESTA-1 for E-selectin targeted delivery.


Subject(s)
Aptamers, Nucleotide/pharmacology , Blood Circulation/drug effects , E-Selectin/metabolism , Endothelial Cells/immunology , Neoplasms/drug therapy , Neoplasms/metabolism , Animals , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/chemistry , Base Sequence , Drug Screening Assays, Antitumor , E-Selectin/chemistry , E-Selectin/genetics , Endothelial Cells/chemistry , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HL-60 Cells , Humans , Kinetics , Ligands , Mice , Molecular Sequence Data , Neoplasms/immunology , Neoplasms/physiopathology , Nucleic Acid Conformation , Phosphates/chemistry , Protein Binding
19.
Biochemistry ; 49(42): 9106-12, 2010 Oct 26.
Article in English | MEDLINE | ID: mdl-20843027

ABSTRACT

CD44, the primary receptor for hyaluronic acid, plays an important role in tumor growth and metastasis. CD44-hyaluronic acid interactions can be exploited for targeted delivery of anticancer agents specifically to cancer cells. Although various splicing variants of CD44 are expressed on the plasma membrane of cancer cells, the hyaluronic acid binding domain (HABD) is highly conserved among the CD44 splicing variants. Using a novel two-step process, we have identified monothiophosphate-modified aptamers (thioaptamers) that specifically bind to the CD44's HABD with high affinities. Binding affinities of the selected thioaptamers for the HABD were in the range of 180-295 nM, an affinity significantly higher than that of hyaluronic acid (K(d) above the micromolar range). The selected thioaptamers bound to CD44 positive human ovarian cancer cell lines (SKOV3, IGROV, and A2780) but failed to bind the CD44 negative NIH3T3 cell line. Our results indicated that thio substitution at specific positions of the DNA phosphate backbone results in specific and high-affinity binding of thioaptamers to CD44. The selected thioaptamers will be of great interest for further development as a targeting or imaging agent for the delivery of therapeutic payloads for cancer tissues.


Subject(s)
Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism , Hyaluronan Receptors/chemistry , Hyaluronan Receptors/metabolism , Hyaluronic Acid/metabolism , Animals , Aptamers, Nucleotide/genetics , Base Sequence , Binding Sites , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Female , Humans , Hyaluronan Receptors/genetics , In Vitro Techniques , Kinetics , Mice , NIH 3T3 Cells , Nucleic Acid Conformation , Ovarian Neoplasms/metabolism , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , SELEX Aptamer Technique
20.
Cancer Res ; 70(9): 3687-96, 2010 May 01.
Article in English | MEDLINE | ID: mdl-20430760

ABSTRACT

RNA interference (RNAi) is a powerful approach for silencing genes associated with a variety of pathologic conditions; however, in vivo RNAi delivery has remained a major challenge due to lack of safe, efficient, and sustained systemic delivery. Here, we report on a novel approach to overcome these limitations using a multistage vector composed of mesoporous silicon particles (stage 1 microparticles, S1MP) loaded with neutral nanoliposomes (dioleoyl phosphatidylcholine, DOPC) containing small interfering RNA (siRNA) targeted against the EphA2 oncoprotein, which is overexpressed in most cancers, including ovarian. Our delivery methods resulted in sustained EphA2 gene silencing for at least 3 weeks in two independent orthotopic mouse models of ovarian cancer following a single i.v. administration of S1MP loaded with EphA2-siRNA-DOPC. Furthermore, a single administration of S1MP loaded with-EphA2-siRNA-DOPC substantially reduced tumor burden, angiogenesis, and cell proliferation compared with a noncoding control siRNA alone (SKOV3ip1, 54%; HeyA8, 57%), with no significant changes in serum chemistries or in proinflammatory cytokines. In summary, we have provided the first in vivo therapeutic validation of a novel, multistage siRNA delivery system for sustained gene silencing with broad applicability to pathologies beyond ovarian neoplasms.


Subject(s)
Nanoparticles/administration & dosage , RNA, Small Interfering/administration & dosage , Silicon/administration & dosage , Animals , Cell Line, Tumor , Female , Gene Silencing , Genetic Therapy/methods , Humans , Liposomes/administration & dosage , Liposomes/chemistry , Liposomes/pharmacokinetics , Mice , Mice, Nude , Nanoparticles/chemistry , Ovarian Neoplasms/genetics , Ovarian Neoplasms/therapy , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/chemistry , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacokinetics , Receptor, EphA2/genetics , Silicon/chemistry , Silicon/pharmacokinetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...