Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
ChemMedChem ; : e202400112, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782722

ABSTRACT

Recent advancements in cancer treatment have improved patient prognoses, but chemotherapy-induced cardiotoxicity remains a prevalent concern. This study explores the potential of F-base-modified aptamers for targeted drug delivery, focusing on their impact on cardiotoxicity. From the phosphoramidite, F-base functionalized Sgc8-F23 was prepared in an automated and programmable way, which was further reacted with Paclitaxel (PTX) to give the F-base modified aptamer Sgc8-paclitaxel conjugates (Sgc8-F23-PTX) efficiently. The conjugate exhibits prolonged circulation time and enhanced efficacy as precision anticancer drug delivery system. Echocardiographic assessments reveal no exacerbation of cardiac dysfunction post-Acute Myocardial Infarction (AMI), and no pathological changes or increased apoptosis in non-infarcted cardiac regions. Autophagy pathway analysis shows no discernible differences in Sgc8-F23-PTX-treated cardiomyocytes compared to controls, contrasting with increased autophagy with Nanoparticle albumin-bound -Paclitaxel (Nab-PTX). Similarly, apoptosis analysis shows no significant distinctions. Moreover, Sgc8-F23-PTX exhibits no inhibitory effects on hERG, hNav1.5, or hCav1.2 channels. These findings suggest the safety and efficacy of F-base-modified Sgc8 aptamers for targeted drug delivery, holding potential clinical applications. Further research is warranted for clinical translation and exploration of other drug carriers.

2.
ACS Appl Mater Interfaces ; 16(12): 14434-14444, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38498684

ABSTRACT

Natural compounds like pterostilbene (PTE) have gained recognition for their various biological activities and potential health benefits. However, challenges related to bioavailability and limited clinical efficacy have prompted efforts to strengthen their therapeutic potential. To meet these challenges, we herein rationally designed and successfully synthesized a pharmaceutical phosphoramidite that allows for the programmable incorporation of PTE into oligonucleotides. The resultant aptamer-PTE conjugate can selectively bind to cancer cells, leading to a specific internalization and drug release. Moreover, compared with free PTE, the conjugate exhibits superior cytotoxicity in cancer cells. Specifically, in a zebrafish xenograft model, the nanomedicine effectively inhibits tumor growth and neovascularization, highlighting its potential for targeted antitumor therapy. This approach presents a promising avenue for harnessing the therapeutic potential of natural compounds via a nanomedicine solution.


Subject(s)
Nanomedicine , Neoplasms , Animals , Humans , Cell Line, Tumor , Neoplasms/drug therapy , Oligonucleotides , Zebrafish
3.
Nano Lett ; 24(3): 983-992, 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38206182

ABSTRACT

On-chip polarization detectors have attracted extensive research interest due to their filterless and ultracompact architecture. However, their polarization-dependent photoresponses cannot be dynamically adjusted, hindering the development toward intelligence. Here, we propose dynamically reconfigurable polarimetry based on in-sensor differentiation of two self-powered photoresponses with orthogonal polarization dependences and tunable responsivities. Such a device can be electrostatically configured in an ultrahigh polarization extinction ratio (PER) mode, where the PER tends to infinity, a Stokes parameter direct sensing mode, where the photoresponse is proportional to S1 or S2 with high accuracy (RMSES1 = 1.5%, RMSES2 = 2.0%), or a background suppressing mode, where the target-background polarization contrast is singularly enhanced. Moreover, the device achieves a polarization angle sensitivity of 0.51 mA·W-1·degree-1 and a specific polarization angle detectivity of 2.8 × 105 cm·Hz1/2·W·degree-1. This scheme is demonstrated throughout the near-to-long-wavelength infrared range, and it will bring a leap for next-generation on-chip polarimeters.

4.
Research (Wash D C) ; 6: 0126, 2023.
Article in English | MEDLINE | ID: mdl-37223462

ABSTRACT

Serving as targeting ligands, aptamers have shown promise in precision medicine. However, the lack of knowledge of the biosafety and metabolism patterns in the human body largely impeded aptamers' clinical translation. To bridge this gap, here we report the first-in-human pharmacokinetics study of protein tyrosine kinase 7 targeted SGC8 aptamer via in vivo PET tracking of gallium-68 (68Ga) radiolabeled aptamers. The specificity and binding affinity of a radiolabeled aptamer, named 68Ga[Ga]-NOTA-SGC8, were maintained as proven in vitro. Further preclinical biosafety and biodistribution evaluation confirmed that aptamers have no biotoxicity, potential mutation risks, or genotoxicity at high dosage (40 mg/kg). Based on this result, a first-in-human clinical trial was approved and carried out to evaluate the circulation and metabolism profiles, as well as biosafety, of the radiolabeled SGC8 aptamer in the human body. Taking advantage of the cutting-edge total-body PET, the aptamers' distribution pattern in the human body was acquired in a dynamic fashion. This study revealed that radiolabeled aptamers are harmless to normal organs and most of them are accumulated in the kidney and cleared from the bladder via urine, which agrees with preclinical studies. Meanwhile, a physiologically based pharmacokinetic model of aptamer was developed, which could potentially predict therapeutic responses and plan personalized treatment strategies. This research studied the biosafety and dynamic pharmacokinetics of aptamers in the human body for the first time, as well as demonstrated the capability of novel molecular imaging fashion in drug development.

5.
Article in English | MEDLINE | ID: mdl-36751121

ABSTRACT

RNA interference (RNAi) mediated by short interfering RNA (siRNA) is a promising method for cancer treatment, but the clinical application is hampered by several limitations, including metabolic instability, lack of tumor specificity, and poor cellular uptake. To meet these challenges, we have explored the possibility of structure modification of siRNA with artificial bases for property optimization. A series of siRNAs functionalized with different numbers of hydrophobic base F are prepared for screening. The interactions of plasma proteins with F-base-modified siRNA (F-siRNA) are investigated, and it is identified that the interaction with serum albumin is dominant. Experiments revealed that the introduction of F bases conferred modified siRNA with improved tumor-specific accumulation, prolonged circulatory retention time, and better tissue permeability. Mechanistic studies indicated that the F base induces the formulation of a stable siRNA-albumin complex, which transports siRNA to tumor tissues selectively owing to an enhanced permeability and retention (EPR) effect of albumin. The F base also facilitates the binding of siRNA to transport-associated proteins on the cell membrane, enabling its cellular internalization. Together, these data demonstrate that F base modification confers siRNA-enhanced cellular uptake and biostability and specific accumulation in tumor tissue, which provides a new approach for the development of siRNA-based cancer therapeutics.

6.
Philos Trans R Soc Lond B Biol Sci ; 378(1871): 20220024, 2023 02 27.
Article in English | MEDLINE | ID: mdl-36633277

ABSTRACT

Classically, a molecular element (ME) is a pure substance composed of two or more atoms of the same element. However, MEs, in the context of this review, can be any molecules as elements bonded together into the backbone of synthetic oligonucleotides (ONs) with designed sequences and functions, including natural A, T, C, G, U, and unnatural bases. The use of MEs can facilitate the synthesis of designer molecules and smart materials. In particular, we discuss the landmarks associated with DNA structure and related technologies, as well as the extensive application of ONs, the ideal type of molecules for intervention therapy aimed at correcting disease-causing genetic errors (indels). It is herein concluded that the discovery of ON therapeutics and the fabrication of designer molecules or nanostructures depend on the ME concept that we previously published. Accordingly, ME will be our focal point as we discuss related research directions and perspectives in making molecules and materials. This article is part of the theme issue 'Reactivity and mechanism in chemical and synthetic biology'.


Subject(s)
DNA , Oligonucleotides , DNA/genetics , DNA/chemistry , Oligonucleotides/chemistry
7.
Front Bioeng Biotechnol ; 10: 1027517, 2022.
Article in English | MEDLINE | ID: mdl-36518194

ABSTRACT

Traditional Chinese Medicine (TCM) provides unique therapeutic effects for many diseases with identified efficacy during long practice. Astragalus Membranaceus (AM) is the Chinese herbal applied for kidney injury in the clinic, but it remains challenging to further enhance the efficacy. Cycloastragenol (CAG) is the ingredient isolated from AM with poor water solubility, which has shown a renoprotective effect. Herein we designed and synthesized the corresponding solid-phase module of CAG, from which CAG as a pharmaceutical element was incorporated into oligonucleotides (ON) as an ON-CAG conjugate in a programmable way by a DNA synthesizer. Cell viability study demonstrated that ON-CAG conjugate remains similar renoprotective effect as that of CAG, which efficiently recovers the activity of HK-2 cells pretreated with cisplatin. Similarly, in the renal cells treated with the conjugate, the biomarkers of kidney injury such as KIM-1 and IL-18 are downregulated, and cytokines are reduced as treated with anti-inflammatory agents. Overall, we have managed to incorporate a hydrophobic ingredient of TCM into ON and demonstrate the oligonucleotide synthesis technology as a unique approach for the mechanism study of TCM, which may facilitate the discovery of new therapeutics based on TCM.

8.
Pharmaceutics ; 14(12)2022 Dec 13.
Article in English | MEDLINE | ID: mdl-36559275

ABSTRACT

Aptamer-drug conjugates (ApDCs) are potential targeted pharmaceutics, but their clinical applications are hampered by fast clearance in blood. Herein we report the construction of ApDCs modified with artificial base F and the study of biological activities. Two types of F-base-modified ApDCs were prepared, Sgc8-paclitaxel by conjugation and Sgc8-gemcitabine, by automated solid-phase synthesis. In vitro experiments showed that F-base-modified ApDCs retain the specificity of the aptamer to target cells and the biological stability is improved. In vivo studies demonstrated that the circulatory time is increased by up to 55 h or longer, as the incorporated F base leads to a stable ApDC-albumin complex as the formulation for targeted delivery. Moreover, conjugated drug molecules were released efficiently and the drug (paclitaxel) concentration in the tumor site was improved. The results demonstrate that an F-base-directed ApDC-albumin complex is a potential platform for drug delivery and targeted cancer therapy.

9.
Molecules ; 27(19)2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36234794

ABSTRACT

Radioiodination of oligonucleotides provides an extra modality for nucleic acid-based theranostics with potential applications. Herein, we report the design and synthesis of a phosphoramidite embedded with a phenolic moiety and demonstrate that oligonucleotides can be readily functionalized with phenol as a precursor by general DNA synthesis. It was identified that the introduction of the precursor does not block the specificity of an aptamer, and the radioiodination is applicable to both DNA and RNA oligonucleotides in a site-specific approach with a commercial kit.


Subject(s)
Nucleic Acids , Oligonucleotides , DNA , Iodine Radioisotopes , Phenols , RNA
10.
Sci Total Environ ; 846: 157381, 2022 Nov 10.
Article in English | MEDLINE | ID: mdl-35850336

ABSTRACT

Estuaries, considered as the important carbon dioxide (CO2), nitrous oxide (N2O) and methane (CH4) sources to the atmosphere, are increasingly affected by near-bottom hypoxia. However, the impact of estuarine hypoxic zone development on GHGs production and discharge remains poorly understood due to the seasonal and spatially distributed heterogeneity of estuarine hypoxia occurrence and the lack of simultaneous monitoring of the distribution of bottom hypoxic waters and the vertical distribution of GHGs. Here, we conducted high spatial resolution vertical stratification sampling and analysis of water column GHGs in the Pearl River Estuary (PRE), a large estuary with frequent hypoxia in recent years. Our results showed that Pearl River runoff is the main source of GHGs in the PRE. Strong nitrification is an important N2O production mechanism in the PRE. In situ generation of water and resuspension of surface sediments were the main sources of CH4 in bottom water, while massive organic matter (OM) mineralization is the main driver of CO2 in bottom water. The development of a hypoxic zone in the PRE significantly increased the concentration of N2O and CH4 in the bottom water and thus increased air-water fluxes. The air-water fluxes of N2O, CH4 and CO2 of PRE in summer were 31.9 ± 7.5 µmol m-2 d-1, 192.5 ± 229.4 µmol m-2 d-1 and 51.9 ± 14.1 mmol m-2 d-1, respectively. This study reveals that GHGs fluxes from estuarine waters to the atmosphere will increase significantly with increasing eutrophication caused by human activities and the expansion of hypoxic zones in estuarine waters.


Subject(s)
Estuaries , Rivers , Carbon Dioxide/analysis , Environmental Monitoring , Humans , Hypoxia , Methane/analysis , Nitrous Oxide/analysis , Seasons , Water/analysis
11.
Angew Chem Int Ed Engl ; 61(31): e202201220, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35536294

ABSTRACT

Aptamers are emerging as promising molecular tools in cancer-targeted theranostics. Improving their in vivo stability has been a critical issue in promoting clinical translation, but such efforts could lead to more serious side effects resulting from prolonged retention in healthy organs. To address this problem, we developed an environment-responsive stabilization strategy for the selective enhancement of aptamer biostability in the tumor microenvironment (TME). Briefly, by means of the end extension of an ATP-responsive protection (ARP) module, the designed aptamer could be protected from nuclease degradation through the specific incorporation of ATP. Based on our in vivo results, this ARP-aptamer probe was effectively accumulated in tumors via aptamer-based molecular recognition. It showed selectively prolonged tumor retention time, but rapid digestion in healthy organs. Our strategy should provide a new paradigm for the development of organ-specific nucleic acid-based imaging and therapeutic agents.


Subject(s)
Aptamers, Nucleotide , Neoplasms , Nucleic Acids , Adenosine Triphosphate , Humans , Neoplasms/drug therapy , Tumor Microenvironment
12.
Nucleic Acids Res ; 50(6): 3083-3095, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35293579

ABSTRACT

Oligonucleotide (ON) therapeutics are emerging as a new generation of medicine with tremendous potential, but their clinical translation is hampered by inferior stability and short circulation time in the human body. Here, we report a general approach to manipulating the interaction between ONs and albumin by modulating hydrophobicity. A series of DNA aptamer derivatives were designed and prepared by programmable synthesis as an ON library with a gradient of hydrophobic base 'F'. In vitro experiments revealed that the introduction of two F bases at both ends of ONs enhanced the biostability without sacrificing biological activities, while the binding affinity toward albumin was dramatically increased with Kd in the range of 100 nM to 1 µM. In vivo imaging confirmed the immediate formation of the aptamer-albumin complex after the injection, and the circulation time of the aptamer was dramatically elongated owing to the enhanced biostability and retarded renal excretion. The programmable incorporation of the F base provides a general approach to regulating albumin-binding affinity and enhancing the stability of aptamers in vivo, conferring aptamer therapeutics prolonged circulation time to meet clinical requirements.


Subject(s)
Aptamers, Nucleotide , Albumins , Aptamers, Nucleotide/chemistry , Humans , SELEX Aptamer Technique
14.
ACS Appl Mater Interfaces ; 13(8): 9457-9463, 2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33356116

ABSTRACT

Converting marketed drug molecules into phosphoramidites may present a potential strategy to facilitate the development of aptamer-drug conjugates (ApDCs) by a DNA synthesizer in a programmable way; however, quite limited methods were reported. Herein, we demonstrated a general approach by repurposing camptothecin (CPT) species. Commonly used inactive ingredients in pharmaceuticals are investigated and selected as a bonding moiety, from which 2-mercaptoethoxy ethanol and thioglycerol were efficiently incorporated with CPT to give the precursors. Cell viability and molecular docking results of the precursors supported that incorporation of the bonding moiety would not interrupt the inhibitory activity. Therefore, corresponding phosphoramidites were prepared as pharmaceutical elements, and a series of ApDCs were constructed automatically by solid-phase synthesis. Biological studies revealed that CPT elements could be specifically delivered to HCT116 cells by an aptamer and released inside cells. This kind of programmable repurposing may take advantage of established safety data and efficacy of existing drugs resulting in a faster development of ApDCs.


Subject(s)
Antineoplastic Agents/pharmacology , Aptamers, Nucleotide/chemistry , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Drug Carriers/chemistry , Topoisomerase I Inhibitors/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/metabolism , Camptothecin/metabolism , DNA/chemistry , DNA Topoisomerases, Type I/metabolism , Drug Design , Drug Repositioning , Drug Screening Assays, Antitumor , HCT116 Cells , Humans , Molecular Docking Simulation , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/metabolism
15.
J Am Chem Soc ; 142(28): 12079-12086, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32516525

ABSTRACT

Antibody drugs have been used to treat many diseases, and to date, this has been the most rapidly growing drug class. However, the lack of suitable methods for real-time and high-throughput monitoring of antibody production and quality control has been a hindrance to the further advancement of antibody drugs or biosimilars. Therefore, we herein report a versatile tool for one-step fluorescence monitoring of antibody production by using aptamer probes selected through the in vitro SELEX method. In this case, DNA aptamers were selected against the humanized IgG1 antibody drug trastuzumab with high specificity and affinity with a Kd value of aptamer CH1S-3 of 10.3 nM. More importantly, the obtained aptamers were able to distinguish native from heat-treated, whereas antibodies failed this test. On the basis of the advantages of rapid detection for aptamers, we designed aptamer molecular beacons for direct and sensitive detection of trastuzumab in complex samples. Unlike traditional antibody-based ELISA, the signal was observed directly upon interaction with the target without the need for time-consuming binding and multiple washing steps. To further highlight biomedical applications, the use of aptamers as potential tools for quality control and traceless purification of antibody drugs was also demonstrated. Thus, aptamers are shown to be promising alternatives for antibody production monitoring, quality control, and purification, providing technical support to accelerate antibody drug development.


Subject(s)
Aptamers, Nucleotide/chemistry , SELEX Aptamer Technique , Trastuzumab/analysis , Antibody Formation , Fluorescence , Quality Control , Trastuzumab/biosynthesis
16.
Anal Chem ; 92(15): 10375-10380, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32527079

ABSTRACT

Artificial bases have emerged as a useful tool to expand genetic alphabets and biomedical applications of oligonucleotides. Herein, we reported that the conformation conversion enhances cellular uptake of hydrophobic 3,5-bis(trifluoromethyl)benzene (F) base double-strand-conjugated oligonucleotides. The formation of the F base double-strand caged the hydrophobic F base in the duplex strand, thus preventing F base from interacting with cells to some extent. However, upon conversion of F base double-strand-conjugated oligonucleotide to F base single-strand-conjugated oligonucleotide, F bases then were allowed to interact with cells by stronger hydrophobic interactions, followed by cellular uptake. The results were concluded as a pairing-induced cage effect of F base and have the potential for the construction of stimuli-responsive cellular uptake of functional nucleic acids.


Subject(s)
Oligonucleotides/chemistry , Oligonucleotides/metabolism , Biological Transport , Carbohydrate Conformation , HeLa Cells , Humans , Molecular Structure , Organophosphorus Compounds/chemical synthesis , Organophosphorus Compounds/chemistry
17.
Bioconjug Chem ; 31(5): 1289-1294, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32223180

ABSTRACT

Bispecific aptamer-drug conjugates (BsApDC) may improve the efficacy of drugs by enhancing cellular internalization and targeted delivery. Nevertheless, the synthesis of single-molecular BsApDC has not yet been reported, and it could be thwarted by synthetic challenges. Herein we report a general approach to synthesize a BsApDC hybridized chemical and biological method. Primers incorporated with 5-Fluorouracil (5-FU), 10-Hydroxycamptothecin, and Maleimidocaproyl-valine-citrulline-p-aminobenzoyloxycarbonyl-monomethyl auristatin E(vcMMAE) were prepared by chemical synthesis, which were converted to corresponding ApDCs efficiently by enzymatic reaction. Biological studies revealed that BsApDC binds with target cells with enhanced internalization and better inhibitory activity, demonstrating the potential of BsApDCs for targeted tumor therapy.


Subject(s)
Aptamers, Nucleotide/chemistry , Pharmaceutical Preparations/chemistry , Aminobenzoates/chemistry , Camptothecin/analogs & derivatives , Camptothecin/chemistry , Cell Line, Tumor , Fluorouracil/chemistry , Humans , Oligopeptides/chemistry
18.
Theranostics ; 10(9): 4030-4041, 2020.
Article in English | MEDLINE | ID: mdl-32226537

ABSTRACT

Rationale: A cascade, or domino, reaction consists of two, or more, consecutive reactions such that subsequent reactions occur only if some chemical functionality has first been established in the prior step. However, while construction of predesigned and desired molecular domino reactors in a tailored manner is a valuable endeavor, it is still challenging. Methods: To address this challenge, we herein report an aptamer-based photodynamic domino reactor built through automated modular synthesis. The engineering of this reactor takes advantage of the well-established solid-phase synthesis platform to incorporate a photosensitizer into G-quadruplex/ hemin DNAzyme at the molecular level. Results: As a proof of concept, our photodynamic domino reactor, termed AS1411/hemin- pyrochlorophyll A, achieves in vivo photodynamic domino reaction for efficient cancer treatment by using a high concentration of hydrogen peroxide (H2O2) in the tumor microenvironment (TME) to produce O2, followed by consecutive generation of singlet oxygen (1O2) using the pre-produced O2. More specifically, phosphoramidite PA (pyrochlorophyll A) is coupled to aptamer AS1411 to form AS1411-PA ApDC able to simultaneously perform in vivo targeted imaging and photodynamic therapy (PDT). The insertion of hemin into the AS1411 G-quadruplex was demonstrated to alleviate tumor hypoxia by decomposition of H2O2 to produce O2. This was followed by the generation of 1O2 by PA to trigger cascading amplified PDT. Conclusion: Therefore, this study provides a general strategy for building an aptamer-based molecular domino reactor through automated modular synthesis. By proof of concept, we further demonstrate a novel method of achieving enhanced PDT, as well as alleviating TME hypoxia at the molecular level.


Subject(s)
Aptamers, Nucleotide , Neoplasms/drug therapy , Oligodeoxyribonucleotides , Photochemotherapy , Photosensitizing Agents , Tumor Hypoxia/drug effects , Tumor Microenvironment/drug effects , Animals , Aptamers, Nucleotide/pharmacology , Aptamers, Nucleotide/therapeutic use , Humans , Hydrogen Peroxide/metabolism , MCF-7 Cells , Mice, Inbred BALB C , Oligodeoxyribonucleotides/pharmacology , Oligodeoxyribonucleotides/therapeutic use , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Singlet Oxygen/metabolism
19.
Bioconjug Chem ; 31(1): 37-42, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31815437

ABSTRACT

Nucleic acid aptamers, also known as "chemical antibodies", have been widely employed in targeted cancer therapy and diagnosis. For example, aptamer-drug conjugates (ApDCs), through covalent conjugation of cytotoxic warheads to aptamers, have demonstrated anticancer efficacy both in vitro and in vivo. However, a general strategy to endow ApDCs with enhanced biostability, prolonged circulation half-life, and high drug loading content remained elusive. Herein, we present a polymeric approach to engineer ApDCs via conjugation of cell-targeting aptamers with water-soluble polyprodrugs containing a reductive environmentally sensitive prodrug and biocompatible brush-like backbone. The resultant high-drug loading Aptamer-PolyproDrug Conjugates (ApPDCs) exhibited high nuclease resistance, extended in vivo circulation time, specific recognition, and cellular uptake to target cells, reduction-triggered and fluorescent-reporting drug release, and effective cytotoxicity. We could also further expand this design principle toward combination therapy by using two kinds of therapeutic drugs with distinct pharmacological mechanisms.


Subject(s)
Antineoplastic Agents/administration & dosage , Aptamers, Nucleotide/chemistry , Drug Carriers/chemistry , Drug Delivery Systems , Neoplasms/drug therapy , Polymers/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Drug Liberation , Humans
20.
Anal Chem ; 91(21): 13818-13823, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31593429

ABSTRACT

Aptamer-drug conjugates (ApDCs) are emerging as targeted therapeutic drugs that can effectively broaden the chemotherapeutic window with higher efficacy and less toxicity. They show promising targeted tumor-killing effects both in vitro and in vivo. However, the mechanisms underlying the cellular internalization and transport of ApDCs remain unclear, and no systematic study on this topic has been reported. Therefore, we herein investigated the endocytic internalization and subsequent transport of ApDCs in mammalian cells through single-particle tracking. We found that ApDC enters the cells mainly by caveolin-mediated endocytosis and that it exhibits cytoskeleton-dependent transport, along microfilaments and microtubules, to acidic endosomes near the cell nucleus in cytoplasm. We also found that the cellular uptake pathways of ApDCs are identical to those of the aptamer itself, confirming that aptamers play a prominent role in the internalization of ApDCs. This study extends our understanding of the internalization and transport process of ApDCs such that the results could serve as the theoretical foundation for designing new ApDCs and, in turn, promoting cancer-targeted therapy.


Subject(s)
Aptamers, Nucleotide/metabolism , Drug Delivery Systems/methods , Endocytosis , Single Molecule Imaging/methods , Animals , Aptamers, Nucleotide/therapeutic use , Biological Transport , Caveolins , Cell Line , Cytoskeleton , Endosomes , Humans , Mammals
SELECTION OF CITATIONS
SEARCH DETAIL
...