Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 127
Filter
Add more filters

Publication year range
1.
Nanomedicine ; 36: 102429, 2021 08.
Article in English | MEDLINE | ID: mdl-34174419

ABSTRACT

Nanoparticles offer targeted delivery of drugs with minimal toxicity to surrounding healthy tissue and have great potential in the management of human papillomavirus (HPV)-related diseases. We synthesized lipid-modified AS1411 aptamers capable of forming nanoaggregates in solution containing Mg2+. The nanoaggregates presented suitable properties for pharmaceutical applications such as small size (100 nm), negative charge, and drug release. The nanoaggregates were loaded with acridine orange derivative C8 for its specific delivery into cervical cancer cell lines and HPV-positive tissue biopsies. This improved inhibition of HeLa proliferation and cell uptake without significantly affecting healthy cells. Finally, the nanoaggregates were incorporated in a gel formulation with promising tissue retention properties aiming at developing a local delivery strategy of the nanoaggregates in the female genital tract. Collectively, these findings suggest that the nanoformulation protocol has great potential for the delivery of both anticancer and antiviral agents, becoming a novel modality for cervical cancer management.


Subject(s)
Antineoplastic Agents , Antiviral Agents , Aptamers, Nucleotide , Cell Proliferation/drug effects , Drug Delivery Systems , Oligodeoxyribonucleotides , Uterine Cervical Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Nucleotide/pharmacology , Female , HeLa Cells , Humans , Oligodeoxyribonucleotides/chemistry , Oligodeoxyribonucleotides/pharmacokinetics , Oligodeoxyribonucleotides/pharmacology , Uterine Cervical Neoplasms/metabolism
2.
Arterioscler Thromb Vasc Biol ; 39(10): 2028-2037, 2019 10.
Article in English | MEDLINE | ID: mdl-31315441

ABSTRACT

OBJECTIVE: Refrigeration-induced binding of VWF (von Willebrand factor) to platelets contributes to the rapid clearance of refrigerated platelets. In this study, we investigate whether inhibiting VWF binding by a DNA-based aptamer ameliorates the clearance of refrigerated platelets without significantly impeding hemostatic functions. Approach and Results: Platelets were refrigerated with or without aptamer ARC1779 for 48 hours. VWF binding, the effective lifetime of ARC1779, platelet post-transfusion recovery and survival, and the hemostatic function were measured. ARC1779 treatment during refrigeration inhibited the platelet-VWF interaction. ARC1779-treated refrigerated murine platelets exhibited increased post-transfusion recovery and survival than untreated ones (recovery of ARC1779-treated platelets: 76.7±5.5%; untreated: 63.7±0.8%; P<0.01. Half-life: 31.4±2.36 hours versus 28.1±0.86 hours; P<0.05). A similar increase was observed for refrigerated human platelets (recovery: 49.4±4.4% versus 36.8±2.1%, P<0.01; half-life: 9.2±1.5 hours versus 8.7±0.9 hours, ns). The effective lifetime of ARC1779 in mice was 2 hours. Additionally, ARC1779 improved the long-term (2 hours after transfusion) hemostatic function of refrigerated platelets (tail bleeding time of mice transfused with ARC1779-treated refrigerated platelets: 160±65 seconds; untreated: 373±96 seconds; P<0.01). The addition of an ARC1779 antidote before transfusion improved the immediate (15 minutes after transfusion) hemostatic function (bleeding time of treated platelets: 149±21 seconds; untreated: 320±36 seconds; P<0.01). CONCLUSIONS: ARC1779 improves the post-transfusion recovery of refrigerated platelets and preserves the long-term hemostatic function of refrigerated platelets. These results suggest that a short-acting inhibitor of the platelet-VWF interaction may be a potential therapeutic option to improve refrigeration of platelets for transfusion treatment.


Subject(s)
Aptamers, Nucleotide/pharmacology , Blood Donors , Blood Platelets/drug effects , Hemostasis/drug effects , Platelet Transfusion , Refrigeration , von Willebrand Factor/metabolism , Animals , Aptamers, Nucleotide/pharmacokinetics , Blood Platelets/metabolism , Cell Survival/drug effects , Female , Half-Life , Humans , Male , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Protein Binding , Time Factors , von Willebrand Factor/genetics
3.
Anal Chem ; 91(13): 8289-8297, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31141341

ABSTRACT

Aptamers, short DNA or RNA oligonucleotides, which evolved from systematic evolution of ligands by exponential enrichment (SELEX), can perform specific target recognition. Papillary thyroid carcinoma (PTC) is of high incidence worldwide, and the prognosis of advanced PTC is poor. Up to now, there is no specific biomarker that can identify PTC and defects still remain in existing diagnostic methods. Here we report an aptamer, termed TC-6, which is generated from tissue-SELEX by using sections of papillary thyroid carcinoma and a normal thyroid gland. TC-6 could specifically target intracellular components of papillary thyroid cells with high affinity ( Kd = 57.66 ± 5.93 nmol/L) and have performed excellent biocompatibility both in vivo and in vitro. Moreover, fluorescence imaging of PTC tumor-bearing mice revealed that TC-6 was able to accumulate in tumor sites and could distinguish thyroid carcinoma from other benign thyroid diseases efficiently. In addition, TC-6d, a truncated aptamer of TC-6, maintained its affinity toward PTC with Kd of 39.20 ± 8.20 nmol/L. Overall, these results indicate that TC-6 is a potential candidate for developing novel tools for diagnosis and targeted therapy of PTC.


Subject(s)
Aptamers, Nucleotide/pharmacokinetics , SELEX Aptamer Technique/methods , Thyroid Cancer, Papillary/diagnosis , Animals , Diagnosis, Differential , Humans , Materials Testing , Mice , Optical Imaging , SELEX Aptamer Technique/standards , Thyroid Cancer, Papillary/diagnostic imaging
4.
Eur J Nucl Med Mol Imaging ; 46(4): 948-956, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30069577

ABSTRACT

OBJECTIVE: Epidermal growth factor receptor (EGFR) is a theranostic biomarker for a variety of cancer types. The aim of the present study was to develop an 18F radiolabeled EGFR targeting RNA aptamer, and to investigate its ability to visualize and quantify EGFR in xenograft models. METHODS: Biolayer interferometry binding assay was used to detect the binding affinity of the alkyne-modified EGFR aptamer MinE07 (denoted as ME07) with recombinant human wild-type EGFR protein and the mutant EGFRvIII protein. Cy5-conjugated ME07 was used for flow cytometry and immunofluorescence staining, and an Alexa Fluor 488-labeled EGFR antibody (ab193244) was used as a control. 18F-Fluorobenzoyl (FB) azide was employed as a synthon to produce 18F-FB-ME07 via click chemistry, and the cellular uptake and internalization characteristics of 18F-FB-ME07 were investigated. Static PET scans, 60-min dynamic scans, and biodistribution study of 18F-FB-ME07 were performed in three types of tumor models. RESULTS: The Kd values of ME07 to wtEGFR and EGFRvIII proteins were 0.3 nM and 271 nM respectively. The A431, U87MG, and HCT-116 cells showed strong, weak, and negative binding with Cy5-ME07, which is consistent with EGFR expression level in these cells. Peak cell uptake values of 18F-FB-ME07 in A431, U87MG and HCT-116 cells were 2.86%, 2.19% and 0.88% of the added dose respectively. The mean internalization of 18F-FB-ME07 in these cells were 60.02%, 53.1%, and 52.8% of the total accumulated radioactivity. In static PET imaging, despite high uptake in the liver and kidneys, 18F-FB-ME07 showed reasonable accumulation in A431 tumors (1.02 ± 0.13 %ID/g at 30 min after injection). Of note, the uptake of 18F-FB-ME07 in A431 xenografts was significantly higher than that in U87MG and HCT-116 xenografts. In A431 xenografted mice, the tumor/blood ratio was 3.89 and the tumor/muscle ratio reached 8.65. CONCLUSIONS: We for the first time generated an aptamer-derived EGFR targeting PET tracer 18F-FB-ME07, which showed highly selective targeting ability in mouse tumor models expressing different levels of EGFR. Our results suggest that 18F-FB-ME07 is a potential EGFR targeting molecular imaging probe for future clinical translation.


Subject(s)
Aptamers, Nucleotide/chemistry , ErbB Receptors/metabolism , Fluorine Radioisotopes , Gene Expression Regulation, Neoplastic , Positron-Emission Tomography , Animals , Aptamers, Nucleotide/pharmacokinetics , Cell Line, Tumor , Female , Humans , Isotope Labeling , Mice , Tissue Distribution
5.
Nanomedicine ; 21: 102061, 2019 10.
Article in English | MEDLINE | ID: mdl-31344499

ABSTRACT

Targeted DNA nanoparticles have been identified as one of the most promising nanocarriers in anti-glioma drug delivery. We established a multifunctional nanosystem for targeted glioma therapy. Tetrahedral framework nucleic acid (tFNA), entering U87MG cells and bEnd.3 cells, was chosen to deliver two aptamers, GMT8 and Gint4.T, and paclitaxel. GMT8 and Gint4.T, which specifically bind with U87MG cells and with PDGFRß, were linked with tFNA, to form Gint4.T-tFNA-GMT8 (GTG). GTG was efficiently internalized by U87MG and bEnd.3 cells and penetrated an in-vitro blood-brain-barrier model. GTG loaded with paclitaxel (GPC) had potentiated anti-glioma efficacy. It inhibited the proliferation, migration, and invasion of U87MG cells, and enhanced apoptosis induction in these cells. The expression of apoptosis-related proteins was significantly changed after treatment with GPC, confirming apoptosis induction. Our study demonstrated that the combination of GTG and paclitaxel has great potential for glioma treatment and tFNA shows great promise for use in drug delivery.


Subject(s)
Aptamers, Nucleotide , Blood-Brain Barrier , Brain Neoplasms , Glioblastoma , Nanoconjugates , Paclitaxel , Animals , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Nucleotide/pharmacology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Glioblastoma/drug therapy , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Nanoconjugates/chemistry , Nanoconjugates/therapeutic use , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Paclitaxel/pharmacology , Rats
6.
Curr Opin Hematol ; 25(5): 382-388, 2018 09.
Article in English | MEDLINE | ID: mdl-30015643

ABSTRACT

PURPOSE OF REVIEW: Since the selection of the first thrombin-binding aptamer in 1992, the use of nucleic acid aptamers to target specific coagulation factors has emerged as a valuable approach for generating novel anticoagulant and procoagulant therapeutics. Herein, we highlight the most recent discoveries involving application of aptamers for those purposes. RECENT FINDINGS: Learning from the successes and pitfalls of the FIXa-targeting aptamer pegnivacogin in preclinical and clinical studies, the latest efforts to develop antidote-controllable anticoagulation strategies for cardiopulmonary bypass that avoid unfractionated heparin involve potentiation of the exosite-binding factor X (FX)a aptamer 11F7t by combination with either a small molecule FXa catalytic site inhibitor or a thrombin aptamer. Recent work has also focused on identifying aptamer inhibitors of contact pathway factors such as FXIa and kallikrein, which may prove to be well tolerated and effective antithrombotic agents in certain clinical settings. Finally, new approaches to develop procoagulant aptamers to control bleeding associated with hemophilia and other coagulopathies involve targeting activated protein C and tissue plasminogen activator. SUMMARY: Overall, these recent findings exemplify the versatility of aptamers to modulate a variety of procoagulant and anticoagulant factors, along with their capacity to be used complementarily with other aptamers or drugs for wide-ranging applications.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Factor IXa , Factor Xa Inhibitors/therapeutic use , Hemostasis , Animals , Antidotes/pharmacokinetics , Antidotes/therapeutic use , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/pharmacokinetics , Cardiopulmonary Bypass , Catalytic Domain , Factor IXa/antagonists & inhibitors , Factor IXa/metabolism , Factor Xa Inhibitors/adverse effects , Factor Xa Inhibitors/pharmacokinetics , Hemophilia A/blood , Hemophilia A/drug therapy , Humans , Kallikreins/metabolism
7.
Med Res Rev ; 37(6): 1518-1539, 2017 11.
Article in English | MEDLINE | ID: mdl-28759115

ABSTRACT

Mucin 1 (MUC1) is a protein usually found on the apical surface of most normal secretory epithelial cells. However, in most adenocarcinomas, MUC1 is overexpressed, so that it not only appears over the entire cell surface, but is also shed as MUC1 fragments into the blood stream. These phenomena pinpoint MUC1 as a potential target for the diagnosis and treatment of cancer; consequently, interest has increased in MUC1 as a molecular target for overcoming cancer therapy challenges. MUC1 currently ranks second among 75 antigen candidates for cancer vaccines, and different antibodies or aptamers against MUC1 protein are proving useful for tracing cancer cells in the emerging field of targeted delivery. The unique properties of MUC1 aptamers as novel targeting agents, and the revolutionary role that MUC1 now plays in cancer therapy, are the focus of this review. Recent advancements in MUC1-targeted cancer therapy are also assessed.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Mucin-1/metabolism , Neoplasms/therapy , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacokinetics , Humans , Mucin-1/genetics , Neoplasms/genetics , Neoplasms/metabolism
8.
Curr Opin Oncol ; 29(5): 366-374, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28692589

ABSTRACT

PURPOSE OF REVIEW: We will describe recently discovered smart aptamers with tumor specificity, with an emphasis on targeted delivery of novel therapeutic molecules, cancer-specific biomarkers, and immunotherapy. RECENT FINDINGS: The development of cancer-specific aptamers has facilitated targeted delivery of potent therapeutic molecules to cancer cells without harming nontumoral cells. This specificity also makes it possible to discover novel cancer biomarkers. Furthermore, alternative immune-checkpoint blockade aptamers have been developed for combinational immunotherapy. SUMMARY: Aptamers selected against cancer cells show cancer specificity, which has great potential for targeting. First, functionalizing targeted aptamers with therapeutic molecule payloads (e.g., small activating RNAs, antimitotic drugs, therapeutic antibodies, and peptides) facilitates successful delivery into cancer cells. This approach greatly improves the therapeutic index by minimizing side-effects in nontumoral cells. Second, cancer-specific proteins have been identified as cancer biomarkers through in-vitro and in-vivo selection, aptamer pull-down assays, and mass spectrometry. These newly discovered biomarkers improve therapeutic intervention and diagnostic specificity. In addition, the development of alternative immune-checkpoint blockade aptamers is suggested for use in combinational immunotherapeutic with current immune blockade regimens, to reduce the resistance and exhaustion of T cells in clinical trials. VIDEO ABSTRACT: http://links.lww.com/COON/A21.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Aptamers, Peptide/therapeutic use , Neoplasms/drug therapy , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Peptide/genetics , Aptamers, Peptide/pharmacokinetics , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/metabolism
9.
Bioorg Med Chem ; 25(3): 1163-1171, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28089349

ABSTRACT

Aptamers are single-stranded oligonucleotides that recognize molecular targets with high affinity and specificity. Aptamer that selectively bind to the protein tyrosine kinase-7 (PTK7) receptor, overexpressed on many cancers, has been labelled as probes for molecular imaging of cancer. Two new PTK7-targeting aptamer probes were developed by coupling frameworks from the fluorescent dye AlexaFluor647 or the 6-hydrazinonicotinamide (HYNIC) chelator-labelled to 99mTc. The derivatizations via a 5'-aminohexyl terminal linker were done at room temperature and under mild buffer conditions. Physicochemical and biological controls for both imaging agents were performed verifying the integrity of the aptamer-conjugates by HPLC. Recognition of melanoma (B16F1) and lymphoma (A20) mouse cell lines by the aptamer was studied using cell binding, flow cytometry and confocal microscopy. Finally, in vivo imaging studies in tumour-bearing mice were performed. The new probes were able to bind to melanoma and lymphoma cell lines in vitro, the in vivo imaging in tumour-bearing mice showed different uptake behaviours showing for the fluorescent conjugate good uptake by B cell lymphoma while the radiolabelled conjugate did not display tumour uptake due to its high extravascular distribution, and both showed rapid clearance properties in tumour-bearing mice.


Subject(s)
Aptamers, Nucleotide/pharmacokinetics , Cell Adhesion Molecules/antagonists & inhibitors , Fluorescent Dyes/pharmacokinetics , Lymphoma/diagnosis , Melanoma/diagnosis , Molecular Imprinting , Protein Kinase Inhibitors/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/chemistry , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Dose-Response Relationship, Drug , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/chemistry , Humans , Molecular Structure , Neoplasms, Experimental/diagnosis , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Structure-Activity Relationship , Tissue Distribution
10.
J Nanobiotechnology ; 15(1): 81, 2017 Nov 13.
Article in English | MEDLINE | ID: mdl-29132385

ABSTRACT

Aptamers are short non-coding, single-stranded oligonucleotides (RNA or DNA) developed through Systematic Evolution of Ligands by Exponential enrichment (SELEX) in vitro. Similar to antibodies, aptamers can bind to specific targets with high affinity, and are considered promising therapeutic agents as they have several advantages over antibodies, including high specificity, stability, and non-immunogenicity. Furthermore, aptamers can be produced at a low cost and easily modified, and are, therefore, called "chemical antibodies". In the past years, a variety of aptamers specifically bound to both breast cancer biomarkers and cells had been selected. Besides, taking advantage of nanomaterials, there were a number of aptamer-nanomaterial conjugates been developed and widely investigated for diagnostics and targeted therapy of breast cancer. In this short review, we first present a systematical review of various aptamer selection methods. Then, various aptamer-based diagnostic and therapeutic strategies of breast cancer were provided. Finally, the current problems, challenges, and future perspectives in the field were thoroughly discussed.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Biomarkers, Tumor/genetics , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Doxorubicin/therapeutic use , SELEX Aptamer Technique , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/therapeutic use , Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/pharmacokinetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/chemistry , Epithelial Cell Adhesion Molecule/genetics , Epithelial Cell Adhesion Molecule/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression , Humans , Ligands , Molecular Targeted Therapy/methods , Nanotubes , Protein Binding , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
11.
Proc Natl Acad Sci U S A ; 111(48): 17099-103, 2014 Dec 02.
Article in English | MEDLINE | ID: mdl-25404344

ABSTRACT

The in vivo application of aptamers as therapeutics could be improved by enhancing target-specific accumulation while minimizing off-target uptake. We designed a light-triggered system that permits spatiotemporal regulation of aptamer activity in vitro and in vivo. Cell binding by the aptamer was prevented by hybridizing the aptamer to a photo-labile complementary oligonucleotide. Upon irradiation at the tumor site, the aptamer was liberated, leading to prolonged intratumoral retention. The relative distribution of the aptamer to the liver and kidney was also significantly decreased, compared to that of the free aptamer.


Subject(s)
Aptamers, Nucleotide/metabolism , Aptamers, Nucleotide/pharmacokinetics , Mammary Neoplasms, Experimental/metabolism , Skin/metabolism , Animals , Aptamers, Nucleotide/genetics , Base Sequence , Carbocyanines/metabolism , Cell Line, Tumor , Fluorescence Resonance Energy Transfer , Mammary Neoplasms, Experimental/pathology , Mice, Nude , Microscopy, Fluorescence , Nucleic Acid Hybridization/radiation effects , Oligonucleotides/genetics , Skin/pathology , Skin/radiation effects , Tissue Distribution , Ultraviolet Rays
12.
Int J Mol Sci ; 18(11)2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29144411

ABSTRACT

Recent progresses in organic chemistry and molecular biology have allowed the emergence of numerous new applications of nucleic acids that markedly deviate from their natural functions. Particularly, DNA and RNA molecules-coined aptamers-can be brought to bind to specific targets with high affinity and selectivity. While aptamers are mainly applied as biosensors, diagnostic agents, tools in proteomics and biotechnology, and as targeted therapeutics, these chemical antibodies slowly begin to be used in other fields. Herein, we review recent progress on the use of aptamers in the construction of smart DNA origami objects and MRI and PET imaging agents. We also describe advances in the use of aptamers in the field of neurosciences (with a particular emphasis on the treatment of neurodegenerative diseases) and as drug delivery systems. Lastly, the use of chemical modifications, modified nucleoside triphosphate particularly, to enhance the binding and stability of aptamers is highlighted.


Subject(s)
Aptamers, Nucleotide/chemistry , Drug Delivery Systems/methods , Nanotechnology/methods , Neuroprotective Agents/administration & dosage , SELEX Aptamer Technique/methods , Animals , Aptamers, Nucleotide/pharmacokinetics , Biosensing Techniques/methods , Humans
13.
RNA Biol ; 13(12): 1232-1245, 2016 12.
Article in English | MEDLINE | ID: mdl-27715478

ABSTRACT

Nucleic acid aptamers are single-stranded DNA or RNA oligonucleotide sequences that bind to a specific target molecule with high affinity and specificity through their ability to adopt 3-dimensional structure in solution. Aptamers have huge potential as targeted therapeutics, diagnostics, delivery agents and as biosensors. However, aptamers composed of natural nucleotide monomers are quickly degraded in vivo and show poor pharmacodynamic properties. To overcome this, chemically-modified nucleic acid aptamers are developed by incorporating modified nucleotides after or during the selection process by Systematic Evolution of Ligands by EXponential enrichment (SELEX). This review will discuss the development of chemically-modified aptamers and provide the pros and cons, and new insights on in vitro aptamer selection strategies by using chemically-modified nucleic acid libraries.


Subject(s)
Aptamers, Nucleotide/chemical synthesis , Aptamers, Nucleotide/pharmacokinetics , Animals , Aptamers, Nucleotide/chemistry , Humans , Nucleic Acid Conformation , SELEX Aptamer Technique , Small Molecule Libraries/chemical synthesis , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacokinetics
14.
Mol Ther ; 22(11): 1910-22, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24954476

ABSTRACT

Cell-targeted therapies (smart drugs), which selectively control cancer cell progression with limited toxicity to normal cells, have been developed to effectively treat some cancers. However, many cancers such as metastatic prostate cancer (PC) have yet to be treated with current smart drug technology. Here, we describe the thorough preclinical characterization of an RNA aptamer (A9g) that functions as a smart drug for PC by inhibiting the enzymatic activity of prostate-specific membrane antigen (PSMA). Treatment of PC cells with A9g results in reduced cell migration/invasion in culture and metastatic disease in vivo. Importantly, A9g is safe in vivo and is not immunogenic in human cells. Pharmacokinetic and biodistribution studies in mice confirm target specificity and absence of non-specific on/off-target effects. In conclusion, these studies provide new and important insights into the role of PSMA in driving carcinogenesis and demonstrate critical endpoints for the translation of a novel RNA smart drug for advanced stage PC.


Subject(s)
Antigens, Surface/metabolism , Aptamers, Nucleotide/administration & dosage , Glutamate Carboxypeptidase II/metabolism , Molecular Targeted Therapy/methods , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/therapy , Animals , Aptamers, Nucleotide/pharmacokinetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Male , Mice , Neoplasm Metastasis , Xenograft Model Antitumor Assays
15.
Nanomedicine ; 11(3): 671-9, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25461281

ABSTRACT

This paper reports an in vivo evaluation of toxicology and biodistribution of a highly anisotropic Au nanoconstruct composed of a gold nanostar (AuNS) core and a ligand shell of a G-quadruplex DNA aptamer AS1411 (Apt) supporting both targeting and therapy capabilities. We examined the toxicity of the nanoconstructs (Apt-AuNS) at four different injected concentrations. At the highest dose tested (48 mg/kg), maximal tolerated dose was not reached. Clinical pathology showed no apparent signs of acute toxicity. Interestingly, the nanoconstructs circulated longer in female rats compared to male rats. In two different tumor models, the biodistribution of Apt-AuNS, especially tumor accumulation, was different. Accumulation of Apt-AuNS was 5 times higher in invasive breast cancer tumors compared to fibrosarcoma tumors. These results provide insight on identifying a tumor model and nanoconstruct for in vivo studies, especially when an in vitro therapeutic response is observed in multiple cancer cell lines. From the clinical editor: This study investigated the toxicity and distribution of aptamer loaded gold nanostars in a rodent model of invasive breast cancer and fibrosarcoma. Acute toxicity was not identified even in the highest studied doses. Fivefold accumulation was demonstrated in the breast cancer model compared to the fibrosarcoma model. Studies like this are critically important in further clarifying the potential therapeutic use of these nanoconstructs, especially when ex vivo effects are clearly demonstrated.


Subject(s)
Aptamers, Nucleotide , Fibrosarcoma/drug therapy , Gold , Mammary Neoplasms, Experimental/drug therapy , Metal Nanoparticles , Animals , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Female , Fibrosarcoma/metabolism , Fibrosarcoma/pathology , Gold/adverse effects , Gold/chemistry , Gold/pharmacokinetics , Gold/pharmacology , Male , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Metal Nanoparticles/adverse effects , Metal Nanoparticles/chemistry , Metal Nanoparticles/therapeutic use , Mice , Mice, Nude , Rats , Sex Characteristics
16.
J Biol Chem ; 288(29): 21136-21147, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23744070

ABSTRACT

Excessive secretion of glucagon, a functional insulin antagonist, significantly contributes to hyperglycemia in type 1 and type 2 diabetes. Accordingly, immunoneutralization of glucagon or genetic deletion of the glucagon receptor improved glucose homeostasis in animal models of diabetes. Despite this strong evidence, agents that selectively interfere with endogenous glucagon have not been implemented in clinical practice yet. We report the discovery of mirror-image DNA-aptamers (Spiegelmer®) that bind and inhibit glucagon. The affinity of the best binding DNA oligonucleotide was remarkably increased (>25-fold) by the introduction of oxygen atoms at selected 2'-positions through deoxyribo- to ribonucleotide exchanges resulting in a mixed DNA/RNA-Spiegelmer (NOX-G15) that binds glucagon with a Kd of 3 nm. NOX-G15 shows no cross-reactivity with related peptides such as glucagon-like peptide-1, glucagon-like peptide-2, gastric-inhibitory peptide, and prepro-vasoactive intestinal peptide. In vitro, NOX-G15 inhibits glucagon-stimulated cAMP production in CHO cells overexpressing the human glucagon receptor with an IC50 of 3.4 nm. A single injection of NOX-G15 ameliorated glucose excursions in intraperitoneal glucose tolerance tests in mice with streptozotocin-induced (type 1) diabetes and in a non-genetic mouse model of type 2 diabetes. In conclusion, the data suggest NOX-G15 as a therapeutic candidate with the potential to acutely attenuate hyperglycemia in type 1 and type 2 diabetes.


Subject(s)
Aptamers, Nucleotide/pharmacology , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 2/blood , Glucagon/antagonists & inhibitors , RNA/metabolism , Animals , Aptamers, Nucleotide/blood , Aptamers, Nucleotide/pharmacokinetics , Aptamers, Nucleotide/therapeutic use , Blood Glucose/metabolism , Body Weight/drug effects , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/biosynthesis , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Disease Models, Animal , Fasting/blood , Glucagon/metabolism , Glucose Tolerance Test , Humans , Kinetics , Male , Mice , Mice, Inbred BALB C
17.
Invest New Drugs ; 32(1): 178-87, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24242861

ABSTRACT

BACKGROUND: DNA aptamers represent a novel strategy in anti-cancer medicine. AS1411, a DNA aptamer targeting nucleolin (a protein which is overexpressed in many tumor types), was evaluated in patients with metastatic, clear-cell, renal cell carcinoma (RCC) who had failed treatment with ≥1 prior tyrosine kinase inhibitor. METHODS: In this phase II, single-arm study, AS1411 was administered at 40 mg/kg/day by continuous intravenous infusion on days 1-4 of a 28-day cycle, for two cycles. Primary endpoint was overall response rate; progression-free survival (PFS) and safety were secondary endpoints. RESULTS: 35 patients were enrolled and treated. One patient (2.9 %) had a response to treatment. The response was dramatic (84 % reduction in tumor burden by RECIST 1.0 criteria) and durable (patient remains free of progression 2 years after completing therapy). Whole exome sequencing of this patient's tumor revealed missense mutations in the mTOR and FGFR2 genes which is of interest because nucleolin is known to upregulate mTOR pathway activity by enhancing AKT1 mRNA translation. No other responses were seen. Thirty-four percent of patients had an AS1411-related adverse event, all of which were mild or moderate. CONCLUSIONS: AS1411 appears to have minimal activity in unselected patients with metastatic RCC. However, rare, dramatic and durable responses can be observed and toxicity is low. One patient in this study had an excellent response and was found to have FGFR2 and mTOR mutations which will be of interest in future efforts to discover and validate predictive biomarkers of response to nucleolin targeted compounds. DNA aptamers represent a novel way to target cancer cells at a molecular level and continue to be developed with a view to improving treatment and imaging in cancer medicine.


Subject(s)
Aptamers, Nucleotide/therapeutic use , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/drug therapy , Oligodeoxyribonucleotides/therapeutic use , Phosphoproteins/antagonists & inhibitors , RNA-Binding Proteins/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Aptamers, Nucleotide/blood , Aptamers, Nucleotide/pharmacokinetics , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/drug therapy , Demography , Exome/genetics , Female , Humans , INDEL Mutation/genetics , Infusions, Intravenous , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Male , Middle Aged , Models, Molecular , Neoplasm Metastasis , Oligodeoxyribonucleotides/blood , Oligodeoxyribonucleotides/pharmacokinetics , Sequence Analysis, DNA , Treatment Outcome , Nucleolin
18.
Mol Ther ; 21(12): 2236-46, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23887360

ABSTRACT

Complement factor C5a is a potent proinflammatory mediator that contributes to the pathogenesis of numerous inflammatory diseases. Here, we describe the discovery of NOX-D20, a PEGylated biostable mirror-image mixed (l-)RNA/DNA aptamer (Spiegelmer) that binds to mouse and human C5a with picomolar affinity. In vitro, NOX-D20 inhibited C5a-induced chemotaxis of a CD88-expressing cell line and efficiently antagonized the activation of primary human polymorphonuclear leukocytes (PMN) by C5a. Binding of NOX-D20 to the C5a moiety of human C5 did not interfere with the formation of the terminal membrane attack complex (MAC). In sepsis, for which a specific interventional therapy is currently lacking, complement activation and elevated levels of C5a are suggested to contribute to multiorgan failure and mortality. In the model of polymicrobial sepsis induced by cecal ligation and puncture (CLP), NOX-D20 attenuated inflammation and organ damage, prevented the breakdown of the vascular endothelial barrier, and improved survival. Our study suggests NOX-D20 as a new therapeutic candidate for the treatment of sepsis.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Aptamers, Nucleotide/therapeutic use , Complement C5a/antagonists & inhibitors , Multiple Organ Failure/prevention & control , Peritonitis/drug therapy , Sepsis/drug therapy , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Aptamers, Nucleotide/pharmacokinetics , Complement C5a/immunology , Complement C5a/metabolism , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Multiple Organ Failure/drug therapy , Neutrophils/immunology , Neutrophils/metabolism , Peritonitis/complications , Peritonitis/physiopathology , Sepsis/complications
19.
Int J Clin Pharmacol Ther ; 52(7): 574-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24755127

ABSTRACT

OBJECTIVE: To rapidly identify patients who will ultimately respond to 1 year of therapy, and optimize their inter dose interval. MATERIALS AND METHODS: An intravitreal (IVT) ophthalmic dosing paradigm was designed based on clinical efficacy, nonclinical pharmacokinetics (PK), and disease progression modeling. Relevant non-clinical PK models were used to extrapolate IVT drug concentrations to patients. RESULTS: Modeling predicted that > 80% of patients who would respond to 1 year of IVT treatment with an improvement in best-corrected visual acuity (BCVA) could be identified after the first 2 doses of treatment. These 2 initial doses produced ~ 75% of the maximum improvement in BCVA attainable. Moreover, the models also predicted those patients who responded after 1 year of treatment may tolerate an extension of the inter dose interval to 12 weeks without significant deterioration of BCVA. In contrast, > 70% of responsive patients who did not respond to 1 year of treatment showed inadequate responses after 2 doses. CONCLUSIONS: These models use data from 2 doses to identify those patients likely to benefit after 1 year of treatment, and thereafter can lengthen their inter dose interval without deleterious effects. This method identifies potential treatment responders early, and lengthens the inter dose interval during long-term administration while allowing non-responders to pursue alternative therapies earlier, thereby minimizing risk to the patient.


Subject(s)
Aptamers, Nucleotide/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aptamers, Nucleotide/adverse effects , Aptamers, Nucleotide/pharmacokinetics , Disease Progression , Humans , Intravitreal Injections , Models, Biological , Visual Acuity/drug effects
20.
Br J Pharmacol ; 181(20): 3869-3885, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38877797

ABSTRACT

BACKGROUND AND PURPOSE: The ability to measure specific molecules at multiple sites within the body simultaneously, and with a time resolution of seconds, could greatly advance our understanding of drug transport and elimination. EXPERIMENTAL APPROACH: As a proof-of-principle demonstration, here we describe the use of electrochemical aptamer-based (EAB) sensors to measure transport of the antibiotic vancomycin from the plasma (measured in the jugular vein) to the cerebrospinal fluid (measured in the lateral ventricle) of live rats with temporal resolution of a few seconds. KEY RESULTS: In our first efforts, we made measurements solely in the ventricle. Doing so we find that, although the collection of hundreds of concentration values over a single drug lifetime enables high-precision estimates of the parameters describing intracranial transport, due to a mathematical equivalence, the data produce two divergent descriptions of the drug's plasma pharmacokinetics that fit the in-brain observations equally well. The simultaneous collection of intravenous measurements, however, resolves this ambiguity, enabling high-precision (typically of ±5 to ±20% at 95% confidence levels) estimates of the key pharmacokinetic parameters describing transport from the blood to the cerebrospinal fluid in individual animals. CONCLUSIONS AND IMPLICATIONS: The availability of simultaneous, high-density 'in-vein' (plasma) and 'in-brain' (cerebrospinal fluid) measurements provides unique opportunities to explore the assumptions almost universally employed in earlier compartmental models of drug transport, allowing the quantitative assessment of, for example, the pharmacokinetic effects of physiological processes such as the bulk transport of the drug out of the CNS via the dural venous sinuses.


Subject(s)
Anti-Bacterial Agents , Vancomycin , Animals , Biological Transport , Male , Rats , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/cerebrospinal fluid , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/administration & dosage , Vancomycin/pharmacokinetics , Vancomycin/cerebrospinal fluid , Vancomycin/blood , Vancomycin/administration & dosage , Rats, Sprague-Dawley , Jugular Veins/metabolism , Brain/metabolism , Aptamers, Nucleotide/pharmacokinetics , Electrochemical Techniques/methods
SELECTION OF CITATIONS
SEARCH DETAIL