Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters








Publication year range
1.
J Drug Target ; 32(5): 510-528, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38512151

ABSTRACT

Aptamers, a novel type of targeted ligand used in drug delivery, have quickly gained popularity due to their high target specificity and affinity. Different aptamer-mediated drug delivery systems, such as aptamer-drug conjugate (ApDC), aptamer-siRNA, and aptamer-functionalised nanoparticle systems, are currently being developed for the successful treatment of cancer based on the excellent properties of aptamers. These systems can decrease potential toxicity and enhance therapeutic efficacy by targeting the drug moiety. In this review, we provide an overview of recent developments in aptamer-mediated delivery systems for cancer therapy, specifically for breast cancer, and talk about the potential applications and current issues of novel aptamer-based techniques. This study in aptamer technology for breast cancer therapy highlights key aptamers targeting well-established biomarkers such as HER2, oestrogen receptor, and progesterone receptor. Additionally, we explore the potential of aptamers in overcoming various challenges such as drug resistance and improving the delivery of therapeutic agents. This review aims to provide a deeper understanding of the present aptamer-based targeted delivery applications through in-depth analysis to increase efficacy and create new therapeutic approaches that may ultimately lead to better treatment outcomes for cancer patients.


Subject(s)
Antineoplastic Agents , Aptamers, Nucleotide , Breast Neoplasms , Drug Delivery Systems , Humans , Breast Neoplasms/drug therapy , Aptamers, Nucleotide/administration & dosage , Female , Drug Delivery Systems/methods , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Animals , Drug Resistance, Neoplasm
2.
Mol Pharm ; 21(4): 1705-1718, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38466144

ABSTRACT

Photodynamic therapy (PDT) is often applied in a clinical setting to treat bladder cancer. However, current photosensitizers report drawbacks such as low efficacy, low selectivity, and numerous side effects, which have limited the clinical values of PDT for bladder cancer. Previously, we developed the first bladder cancer-specific aptamer that can selectively bind to and be internalized by bladder tumor cells versus normal uroepithelium cells. Here, we use an aptamer-based drug delivery system to deliver photosensitizer chlorine e6 (Ce6) into bladder tumor cells. In addition to Ce6, we also incorporate catalase into the drug complex to increase local oxygen levels in the tumor tissue. Compared with free Ce6, an aptamer-guided DNA nanotrain (NT) loaded with Ce6 and catalase (NT-Catalase-Ce6) can specifically recognize bladder cancer cells, produce oxygen locally, induce ROS in tumor cells, and cause mitochondrial apoptosis. In an orthotopic mouse model of bladder cancer, the intravesical instillation of NT-Catalase-Ce6 exhibits faster drug internalization and a longer drug retention time in tumor tissue compared with that in normal urothelium. Moreover, our modified PDT significantly inhibits tumor growth with fewer side effects such as cystitis than free Ce6. This aptamer-based photosensitizer delivery system can therefore improve the selectivity and efficacy and reduce the side effects of PDT treatment in mouse models of bladder cancer, bearing a great translational value for bladder cancer intravesical therapy.


Subject(s)
Chlorophyllides , Photochemotherapy , Porphyrins , Urinary Bladder Neoplasms , Animals , Mice , Catalase/therapeutic use , Cell Line, Tumor , Oxygen , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Humans
4.
Int J Biol Macromol ; 238: 124173, 2023 May 31.
Article in English | MEDLINE | ID: mdl-36965552

ABSTRACT

Aptamers with high affinity and specificity for certain targets have rapidly become a novel class of targeted ligands applicated in drug delivery. Based on the excellent characteristics of aptamers, different aptamer-mediated drug delivery systems have been developed, including aptamer-drug conjugate (ApDC), aptamer-siRNA, and aptamer-functionalized nanoparticle systems for the effective treatment of cancer, which can reduce potential toxicity and improve therapeutic efficacy. In this review, we summarize the recent progress of aptamer-mediated delivery systems in cancer therapy, and discuss the application prospects and existing problems of innovative approaches based on aptamer therapy. Overall, this review aims to better understand the current aptamer-based targeted delivery applications through in-depth analysis to improve efficacy and develop new therapeutic methods which can ultimately improve treatment outcomes for cancer patients.


Subject(s)
Aptamers, Nucleotide , Nanoparticles , Neoplasms , Humans , Drug Delivery Systems/methods , Neoplasms/drug therapy , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use
5.
Biosens Bioelectron ; 218: 114763, 2022 Dec 15.
Article in English | MEDLINE | ID: mdl-36240628

ABSTRACT

Mitochondrion has emerged as a promising drug target for photodynamic therapy (PDT), due to its significant role in supporting life activities and being reactive oxygen species (ROS)-sensitive. Herein, we establish a new strategy that in-situ bio-synthesized Au NCs combine with mitochondria-targeted aptamer-Pyro conjugates (ApPCs) for specific tumor imaging and PDT. The prepared ApPCs can serve as template for the in-situ bio-synthesis of Au NCs, thereby facilitating the generation of Au NCs-ApPCs assemblies in unique tumor microenvironment. Compared with highly negatively charged ApPCs, bio-synthesized nanoscale Au NCs-ApPCs assemblies are conducive to cell uptake, which consequently benefits the delivery of ApPCs. After dissociated from Au NCs-ApPCs, internalized ApPCs can selectively accumulate in mitochondria and generate excess ROS to disrupt the mitochondrial membrane upon irradiation, thus inducing efficient cell killing. In vitro assays demonstrated that the fluorescent Au NCs-ApPCs assemblies could be specifically produced in cancerous cells, indicating the specific tumor imaging ability, while intracellular ApPCs co-localized well with mitochondria. CCK-8 results revealed over 80% cell death after PDT. In vivo study showed that fluorescent Au NCs-ApPCs assemblies were exclusively generated in tumor and achieved long-term retention; tumor growth was significantly inhibited after 15-day PDT treatment. All these evidences suggest that in-situ bio-synthesized Au NCs-ApPCs assembly is a potent mitochondria-targeted nanoprobe to boost the PDT efficacy of cancers.


Subject(s)
Biosensing Techniques , Metal Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Gold , Reactive Oxygen Species/metabolism , Sincalide , Mitochondria/metabolism , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Tumor Microenvironment
6.
Molecules ; 27(19)2022 Oct 06.
Article in English | MEDLINE | ID: mdl-36235168

ABSTRACT

Cancer is one of the top leading causes of death worldwide. It is a heterogenous disease characterized by unregulated cell proliferation and invasiveness of abnormal cells. For the treatment of cancer, natural products have been widely used as a source of therapeutic ingredients since ancient times. Although natural compounds and their derivatives have demonstrated strong antitumor activity in many types of cancer, their poor pharmacokinetic properties, low cell selectivity, limited bioavailability and restricted efficacy against drug-resistant cancer cells hinder their wide clinical application. Conjugation of natural products with other bioactive molecules has given rise to a new field in drug discovery resulting to the development of novel, bifunctional and more potent drugs for cancer therapy to overcome the current drawbacks. This review discusses multiple categories of such bifunctional conjugates and highlights recent trends and advances in the development of natural product hybrids. Among them, ADCs, PDCs, ApDCs, PROTACs and AUTOTACs represent emerging therapeutic agents against cancer.


Subject(s)
Antineoplastic Agents , Biological Products , Immunoconjugates , Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biological Products/pharmacology , Biological Products/therapeutic use , Cell Proliferation , Humans , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Neoplasms/metabolism
7.
Eur J Med Chem ; 236: 114335, 2022 Jun 05.
Article in English | MEDLINE | ID: mdl-35398732

ABSTRACT

Targeted therapy showed broad application prospects in the treatment of various types of cancer. Through carriers such as aptamers, antibodies, proteins and peptides, targeted therapy can selectively deliver drugs into tumor cells. Compared with traditional treatment methods such as chemo- and radiotherapy, targeted drug delivery systems can reduce the toxic effects of drugs on normal cells and avoid adverse reactions. Herein, an aptamer-cyclometalated iridium(III) complex conjugate (ApIrC) has been designed and developed as a targeted anticancer agent. Owing to the targeting ability of aptamers, ApIrC specifically bound to nucleolin over-expressed on the surface of cancer cells and showed strong fluorescence signal for tumor imaging and diagnosis. ApIrC had more substantial cellular uptake in cancer cells than the iridium complex alone and exhibited favorable low toxicity to normal cells. After uptake by cells through endocytosis, ApIrC can selectively accumulated in mitochondria and induced caspase-3/7-dependent cell death. Remarkably, ApIrC can also specifically target 3D multicellular spheroids (MCSs) and show excellent tumor permeability. So, it can effectively reach the interior of MCSs and cause cell damage. To our knowledge, this is the first report of the aptamer-cyclometalated iridium(III) complex conjugate which studied for cancer targeted therapy. The developed conjugate has great potential to be developed as novel therapeutics for effective and low-toxic cancer treatment.


Subject(s)
Antineoplastic Agents , Aptamers, Nucleotide , Neoplasms , Aptamers, Nucleotide/pharmacology , Cell Line, Tumor , Drug Delivery Systems/methods , Iridium/pharmacology , Mitochondria , Neoplasms/drug therapy
8.
Mol Ther Nucleic Acids ; 27: 1249-1259, 2022 Mar 08.
Article in English | MEDLINE | ID: mdl-35282414

ABSTRACT

Aptamers have excellent specificity and affinity in targeting cell surface receptors, showing great potential in targeted delivery of drugs, siRNA, mRNA, and various nanomaterials with therapeutic function. A better insight of the receptor-mediated internalization process of aptameric conjugates could facilitate the design of new targeted drugs. In this paper, human transferrin receptor-targeted DNA aptamer (termed HG1-9)-fluorophore conjugates were synthesized to visualize the internalization, intracellular transport, and nano-environmental pH of aptameric conjugates. Unlike transferrin that showed high recycling rate and short duration time in cells, the synthetic aptameric conjugates continuously accumulated within cells at a relatively slower rate, besides recycling back to cell surface. After long incubation (≥2 h), only very small amounts of HG1-9 conjugates (approximately 5%) entered late endosomes or lysosomes, and more than 90% of internalized HG1-9 was retained in cellular vesicles (pH 6.0-6.8), escaping from degradation. And among the internalized HG1-9 conjugates, approximately 20% was dissociated from transferrin receptor. The lower recycling ratios of HG1-9 conjugates and their dissociation from receptors promote the accurate and efficient release of their loaded drugs. These results suggest that aptamer HG1-9 could be provided as a versatile tool for specific and effective delivery of diverse therapeutic payloads.

9.
ACS Appl Mater Interfaces ; 14(1): 32-40, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-34928139

ABSTRACT

Uveal melanoma (UM) is the most common primary intraocular malignancy among adults. Despite significant advances in diagnosis and treatment, the general mortality of UM remains alarmingly high. This calls for the development of new approaches for the treatment of UM, such as targeted cancer therapy. CD71, also known as transferrin receptor 1, is overexpressed in UM cell lines and tissues. Herein, we report the development of a CD71-specific aptamer targeting the XQ-2d-MMAE conjugate that can distinguish UM cells from normal human uveal melanocytes. The cytotoxic drug monomethyl auristatin E (MMAE) could be easily coupled onto XQ-2d, a DNA aptamer that specifically targets CD71, to achieve efficiently targeted cancer growth inhibition in a mouse xenograft model, thus implying that XQ-2d-MMAE might be developed into a promising novel anti-tumor agent for the treatment of UM. Collectively, our results demonstrated that CD71 is a reliable target for drug delivery in UM and could be utilized as a model to explore aptamer-mediated targeted UM treatment strategies.


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Melanoma/drug therapy , Oligopeptides/therapeutic use , Uveal Neoplasms/drug therapy , Animals , Antigens, CD/metabolism , Antineoplastic Agents/metabolism , Aptamers, Nucleotide/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Melanoma/metabolism , Mice, Nude , Receptors, Transferrin/metabolism , Uveal Neoplasms/metabolism , Xenograft Model Antitumor Assays
10.
Pharmaceutics ; 13(8)2021 Aug 07.
Article in English | MEDLINE | ID: mdl-34452182

ABSTRACT

Doxorubicin (DOX) is a common anti-tumor drug that binds to DNA or RNA via non-covalent intercalation between G-C sequences. As a therapeutic agent, DOX has been used to form aptamer-drug conjugates for targeted cancer therapy in vitro and in vivo. To improve the therapeutic potential of aptamer-DOX conjugates, we synthesized trifurcated Newkome-type monomer (TNM) structures with three DOX molecules bound through pH-sensitive hydrazone bonds to formulate TNM-DOX. The aptamer-TNM-DOX conjugate (Apt-TNM-DOX) was produced through a simple self-loading process. Chemical validation revealed that Apt-TNM-DOX stably carried high drug payloads of 15 DOX molecules per aptamer sequence. Functional characterization showed that DOX payload release from Apt-TNM-DOX was pH-dependent and occurred at pH 5.0, which reflects the microenvironment of tumor cell lysosomes. Further, Apt-TNM-DOX specifically targeted lymphoma cells without affecting off-target control cells. Aptamer-mediated cell binding resulted in the uptake of Apt-TNM-DOX into targeted cells and the release of DOX payload within cell lysosomes to inhibit growth of targeted lymphoma cells. The Apt-TNM-DOX provides a simple, non-toxic approach to develop aptamer-based targeted therapeutics and may reduce the non-specific side effects associated with traditional chemotherapy.

11.
Cancers (Basel) ; 12(11)2020 Oct 31.
Article in English | MEDLINE | ID: mdl-33142831

ABSTRACT

Recent advances in chemotherapy treatments are increasingly targeted therapies, with the drug conjugated to an antibody able to deliver it directly to the tumor. As high-affinity chemical ligands that are much smaller in size, aptamers are ideal for this type of drug targeting. Aptamer-highly toxic drug conjugates (ApTDCs) based on the E3 aptamer, selected on prostate cancer cells, target and inhibit prostate tumor growth in vivo. Here, we observe that E3 also broadly targets numerous other cancer types, apparently representing a universal aptamer for cancer targeting. Accordingly, ApTDCs formed by conjugation of E3 to the drugs monomethyl auristatin E (MMAE) or monomethyl auristatin F (MMAF) efficiently target and kill a range of different cancer cells. Notably, this targeting extends to both patient-derived explant (PDX) cancer cell lines and tumors, with the E3 MMAE and MMAF conjugates inhibiting PDX cell growth in vitro and with the E3 aptamer targeting PDX colorectal tumors in vivo.

12.
Pharmaceutics ; 12(10)2020 Oct 18.
Article in English | MEDLINE | ID: mdl-33080969

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common malignancy of the liver, which can progress rapidly and has a poor prognosis. Glypican-3 (GPC3) has been proposed to be an important diagnostic biomarker and therapeutic target for HCC. Aptamers have emerged as promising drug delivery vehicles because of their high binding affinity for target molecules. Herein, we developed G12msi, a gemcitabine-incorporated DNA aptamer, targeting GPC3, and evaluated its binding specificity and anti-tumor efficacy in GPC3-overexpressing HCC cell lines and murine xenograft models. GPC3-targeted aptamers were selected by using the SELEX process and the chemotherapy drug gemcitabine was internally incorporated into the aptamer. To determine the binding affinity and internalization of the G12msi, flow cytometry and confocal microscopy were performed on GPC3-positive HepG2, Hep3B, and Huh7 cells, as well as a GPC3-negative A431 cell. The anti-tumor activities of G12msi were evaluated with in vitro and in vivo models. We found that G12msi binds to GPC3-overexpressing HCC tumor cells with high specificity and is effectively internalized. Moreover, G12msi treatment inhibited the cell proliferation of GPC3-positive HCC cell lines with minimal cytotoxicity in control A431 cells. In vivo systemic administration of G12msi significantly inhibited tumor growth of HCC HepG2 cells in xenograft models without causing toxicity. These results suggest that gemcitabine-incorporated GPC3 aptamer-based drug delivery may be a promising strategy for the treatment of HCC.

13.
Mol Ther Nucleic Acids ; 21: 1074-1086, 2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32854062

ABSTRACT

Aptamers are small, functional single-stranded DNA or RNA oligonucleotides that bind to their targets with high affinity and specificity. Experimentally, aptamers are selected by the systematic evolution of ligands by exponential enrichment (SELEX) method. Here, we have used rational drug designing and bioinformatics methods to design the aptamers, which involves three different steps. First, finding a probable aptamer-binding site, and second, designing the recognition and structural parts of the aptamers by generating a virtual library of sequences, selection of specific sequence via molecular docking, molecular dynamics (MD) simulation, binding energy calculations, and finally evaluating the experimental affinity. Following this strategy, a 16-mer DNA aptamer was designed for Annexin A1 (ANXA1). In a direct binding assay, DNA1 aptamer bound to the ANXA1 with dissociation constants value of 83 nM. Flow cytometry and fluorescence microscopy results also showed that DNA1 aptamer binds specifically to A549, HepG2, U-87 MG cancer cells that overexpress ANXA1 protein, but not to MCF7 and L-02, which are ANXA1 negative cells. We further developed a novel system by conjugating DNA1 aptamer with doxorubicin and its efficacy was studied by cellular uptake and cell viability assay. Also, anti-tumor analysis showed that conjugation of doxorubicin with aptamer significantly enhances targeted therapy against tumors while minimizing overall adverse effects on mice health.

14.
J Control Release ; 323: 530-548, 2020 07 10.
Article in English | MEDLINE | ID: mdl-32380206

ABSTRACT

Aptamers are a class of folded nucleic acid strands capable of binding to different target molecules with high affinity and selectivity. Over the years, they have gained a substantial amount of interest as promising molecular tools for numerous medical applications, particularly in targeted therapeutics. However, only the different treatment approaches and current developments of aptamer-drug therapies have been discussed so far, ignoring the crucial technical and functional aspects of constructing a therapeutically effective aptamer-driven drug delivery system that translates to improved in-vivo performance. Hence, this paper provides a comprehensive review of the strategies used to improve the therapeutic performance of aptamer-guided delivery systems. We focus on the different functional features such as drug deployment, payload capacity, in-vivo stability and targeting efficiency to further our knowledge in enhancing the cell-specific delivery of aptamer-drug conjugates. Each reported strategy is critically discussed to emphasize both the benefits provided in comparison with other similar techniques and to outline their potential drawbacks with respect to the molecular properties of the aptamers, the drug and the system to be designed. The molecular architecture and design considerations for an efficient aptamer-based delivery system are also briefly elaborated.


Subject(s)
Antineoplastic Agents , Aptamers, Nucleotide , Neoplasms , Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Drug Delivery Systems , Humans , Neoplasms/drug therapy
15.
Angew Chem Int Ed Engl ; 58(5): 1437-1441, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30537284

ABSTRACT

Combination chemotherapy must strike a difficult balance between safety and efficacy. Current regimens suffer from poor therapeutic impact because drugs are given at their maximum tolerated dose (MTD), which compounds the toxicity risk and exposes tumors to non-optimal drug ratios. A modular framework has been developed that selectively delivers drug combinations at synergistic ratios via tumor-targeting aptamers for effective low-dose treatment. A nucleolin-recognizing aptamer was coupled to peptide scaffolds laden with precise ratios of doxorubicin (DOX) and camptothecin (CPT). This construct had an extremely low IC50 (31.9 nm) against MDA-MB-231 breast cancer cells in vitro, and exhibited in vivo efficacy at micro-dose injections (500 and 350 µg kg-1 dose-1 of DOX and CPT, respectively) that are 20-30-fold lower than their previously-reported MTDs. This approach represents a generalizable strategy for the safe and consistent delivery of combination drugs in oncology.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Aptamers, Nucleotide/chemistry , Camptothecin/therapeutic use , Doxorubicin/therapeutic use , Neoplasms/drug therapy , Peptides/chemistry , Antineoplastic Combined Chemotherapy Protocols/chemistry , Camptothecin/chemistry , Cell Line , Cell Proliferation/drug effects , Doxorubicin/chemistry , Drug Screening Assays, Antitumor , Humans , Maximum Tolerated Dose , Molecular Structure , Neoplasms/pathology
16.
Adv Drug Deliv Rev ; 134: 65-78, 2018 09.
Article in English | MEDLINE | ID: mdl-30125604

ABSTRACT

Precision medicine holds great promise to harness genetic and epigenetic cues for targeted treatment of a variety of diseases, ranging from many types of cancers, neurodegenerative diseases, to cardiovascular diseases. The proteomic profiles resulting from the unique genetic and epigenetic signatures represent a class of relatively well accessible molecular targets for both interrogation (e.g., diagnosis, prognosis) and intervention (e.g., targeted therapy) of these diseases. Aptamers are promising for such applications by specific binding with cognate disease biomarkers. Nucleic acid aptamers are a class of DNA or RNA with unique three-dimensional conformations that allow them to specifically bind with target molecules. Aptamers can be relatively easily screened, reproducibly manufactured, programmably designed, and chemically modified for various biomedical applications, including targeted therapy. Aptamers can be chemically modified to resist enzymatic degradation or optimize their pharmacological behaviors, which ensured their chemical integrity and bioavailability under physiological conditions. In this review, we will focus on recent progress and discuss the challenges and opportunities in the research areas of aptamer-based targeted therapy in the forms of aptamer therapeutics and aptamer-drug conjugates (ApDCs).


Subject(s)
Antineoplastic Agents/therapeutic use , Aptamers, Nucleotide/therapeutic use , Cardiovascular Diseases/drug therapy , Neoplasms/drug therapy , Neurodegenerative Diseases/drug therapy , Neuroprotective Agents/therapeutic use , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Drug Delivery Systems , Humans , Neuroprotective Agents/chemistry , Precision Medicine
17.
Proc Natl Acad Sci U S A ; 115(18): 4761-4766, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29666232

ABSTRACT

Therapies that can eliminate both local and metastatic prostate tumor lesions while sparing normal organ tissue are desperately needed. With the goal of developing an improved drug-targeting strategy, we turned to a new class of targeted anticancer therapeutics: aptamers conjugated to highly toxic chemotherapeutics. Cell selection for aptamers with prostate cancer specificity yielded the E3 aptamer, which internalizes into prostate cancer cells without targeting normal prostate cells. Chemical conjugation of E3 to the drugs monomethyl auristatin E (MMAE) and monomethyl auristatin F (MMAF) yields a potent cytotoxic agent that efficiently kills prostate cancer cells in vitro but does not affect normal prostate epithelial cells. Importantly, the E3 aptamer targets tumors in vivo and treatment with the MMAF-E3 conjugate significantly inhibits prostate cancer growth in mice, demonstrating the in vivo utility of aptamer-drug conjugates. Additionally, we report the use of antidotes to block E3 aptamer-drug conjugate cytotoxicity, providing a safety switch in the unexpected event of normal cell killing in vivo.


Subject(s)
Aminobenzoates/pharmacology , Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/pharmacology , Oligopeptides/pharmacology , Prostatic Neoplasms/drug therapy , Aminobenzoates/chemistry , Animals , Antineoplastic Agents/chemistry , Aptamers, Nucleotide/chemistry , Delayed-Action Preparations/chemistry , Delayed-Action Preparations/pharmacology , Humans , Male , Mice , Mice, Nude , Oligopeptides/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
18.
Int J Nanomedicine ; 13: 763-776, 2018.
Article in English | MEDLINE | ID: mdl-29440899

ABSTRACT

INTRODUCTION: The toxic side effects of doxorubicin (DOX) have limited its use in chemotherapy. Neither liposomal DOX nor pegylated liposomal DOX are able to completely resolve this issue. This is a proof-of-concept study testing aptamer-drug conjugate (ApDC) targeted delivery systems for chemotherapeutic drugs. METHODS: Aptamer library targeting human epidermal growth factor receptor 3 (HER3) was screened and affinity was determined by enzyme-linked immunosorbent assay. Specificity was tested in MCF-7HER3-high, BT474HER3-high, and 293THER3-negative cells using flow cytometry and confocal microscopy. We further developed a HER3 aptamer-functionalized liposome encapsulating DOX and the efficiency of this ApDC was detected by cellular uptake analysis and cell viability assay. In MCF-7 tumor-bearing mice, tumor targeting evaluation, efficacy, toxicity and preliminary pharmocokinetic study was performed. RESULTS: The candidate #13 aptamer had highest affinity (Kd =98±9.7 nM) and specificity. ApDC effectively reduces the half maximal inhibitory concentration of DOX compared with lipsome-DOX and free DOX. In vivo imaging and preliminary distribution studies showed that actively targeted nanoparticles, such as Apt-Lip-DOX molecules, could facilitate the delivery of DOX into tumors in MCF-7-bearing mice. This targeted chemotherapy caused greater tumor suppression than other groups and alleviated side effects such as weight loss, low survival rate, and organ (heart and liver) injury demonstrated by H&E staining. CONCLUSION: The results indicate that targeted chemotherapy using the aptamer-drug conjugate format could provide better tolerability and efficacy compared with non-targeted delivery in relatively low-dose toxic drugs.


Subject(s)
Aptamers, Nucleotide/metabolism , Cardiotoxicity/etiology , Doxorubicin/administration & dosage , Drug Delivery Systems/methods , Nanoparticles/administration & dosage , Receptor, ErbB-3/metabolism , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacokinetics , Cell Line, Tumor , Cell Survival/drug effects , Doxorubicin/adverse effects , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Enzyme-Linked Immunosorbent Assay , Female , Humans , Liposomes/chemistry , MCF-7 Cells , Mice, Inbred BALB C , Nanoparticles/chemistry , Nanoparticles/toxicity , Polyethylene Glycols/administration & dosage , Polyethylene Glycols/adverse effects , Tissue Distribution , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL