Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Br J Ophthalmol ; 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38839251

ABSTRACT

BACKGROUND/AIMS: The aim of this study was to develop and evaluate digital ray, based on preoperative and postoperative image pairs using style transfer generative adversarial networks (GANs), to enhance cataractous fundus images for improved retinopathy detection. METHODS: For eligible cataract patients, preoperative and postoperative colour fundus photographs (CFP) and ultra-wide field (UWF) images were captured. Then, both the original CycleGAN and a modified CycleGAN (C2ycleGAN) framework were adopted for image generation and quantitatively compared using Frechet Inception Distance (FID) and Kernel Inception Distance (KID). Additionally, CFP and UWF images from another cataract cohort were used to test model performances. Different panels of ophthalmologists evaluated the quality, authenticity and diagnostic efficacy of the generated images. RESULTS: A total of 959 CFP and 1009 UWF image pairs were included in model development. FID and KID indicated that images generated by C2ycleGAN presented significantly improved quality. Based on ophthalmologists' average ratings, the percentages of inadequate-quality images decreased from 32% to 18.8% for CFP, and from 18.7% to 14.7% for UWF. Only 24.8% and 13.8% of generated CFP and UWF images could be recognised as synthetic. The accuracy of retinopathy detection significantly increased from 78% to 91% for CFP and from 91% to 93% for UWF. For retinopathy subtype diagnosis, the accuracies also increased from 87%-94% to 91%-100% for CFP and from 87%-95% to 93%-97% for UWF. CONCLUSION: Digital ray could generate realistic postoperative CFP and UWF images with enhanced quality and accuracy for overall detection and subtype diagnosis of retinopathies, especially for CFP.\ TRIAL REGISTRATION NUMBER: This study was registered with ClinicalTrials.gov (NCT05491798).

2.
Adv Healthc Mater ; 13(15): e2304130, 2024 06.
Article in English | MEDLINE | ID: mdl-38427696

ABSTRACT

P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) often leads to the failure of antitumor chemotherapy, and codelivery of chemodrug with P-gp siRNA (siP-gp) represents a promising approach for treating chemoresistant tumors. To maximize the antitumor efficacy, it is desired that the chemodrug be latently released upon completion of siP-gp-mediated gene silencing, which however, largely remains an unmet demand. Herein, core-shell nanocomplexes (NCs) are developed to overcome MDR via staged liberation of siP-gp and chemodrug (doxorubicin, Dox) in hierarchical response to reactive oxygen species (ROS) concentration gradients. The NCs are constructed from mesoporous silica nanoparticles (MSNs) surface-decorated with cRGD-modified, PEGylated, ditellurium-crosslinked polyethylenimine (RPPT), wherein thioketal-linked dimeric doxorubicin (TK-Dox2) and photosensitizer are coencapsulated inside MSNs while siP-gp is embedded in the RPPT polymeric layer. RPPT with ultrahigh ROS-sensitivity can be efficiently degraded by the low-concentration ROS inside cancer cells to trigger siP-gp release. Upon siP-gp-mediated gene silencing and MDR reversal, light irradiation is performed to generate high-concentration, lethal amount of ROS, which cleaves thioketal with low ROS-sensitivity to liberate the monomeric Dox. Such a latent release profile greatly enhances Dox accumulation in Dox-resistant cancer cells (MCF-7/ADR) in vitro and in vivo, which cooperates with the generated ROS to efficiently eradicate MCF-7/ADR xenograft tumors.


Subject(s)
Doxorubicin , Drug Resistance, Neoplasm , Nanoparticles , RNA, Small Interfering , Reactive Oxygen Species , Humans , Reactive Oxygen Species/metabolism , Doxorubicin/pharmacology , Doxorubicin/chemistry , RNA, Small Interfering/chemistry , Drug Resistance, Neoplasm/drug effects , Animals , Nanoparticles/chemistry , Mice , Mice, Nude , Female , Silicon Dioxide/chemistry , Cell Line, Tumor , MCF-7 Cells , Mice, Inbred BALB C , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Drug Resistance, Multiple/drug effects , Photosensitizing Agents/chemistry , Photosensitizing Agents/pharmacology
3.
Proc Natl Acad Sci U S A ; 120(36): e2302342120, 2023 09 05.
Article in English | MEDLINE | ID: mdl-37639589

ABSTRACT

Inhibition of overexpressed enzymes is among the most promising approaches for targeted cancer treatment. However, many cancer-expressed enzymes are "nonlethal," in that the inhibition of the enzymes' activity is insufficient to kill cancer cells. Conventional antibody-based therapeutics can mediate efficient treatment by targeting extracellular nonlethal targets but can hardly target intracellular enzymes. Herein, we report a cancer targeting and treatment strategy to utilize intracellular nonlethal enzymes through a combination of selective cancer stem-like cell (CSC) labeling and Click chemistry-mediated drug delivery. A de novo designed compound, AAMCHO [N-(3,4,6-triacetyl- N-azidoacetylmannosamine)-cis-2-ethyl-3-formylacrylamideglycoside], selectively labeled cancer CSCs in vitro and in vivo through enzymatic oxidation by intracellular aldehyde dehydrogenase 1A1. Notably, azide labeling is more efficient in identifying tumorigenic cell populations than endogenous markers such as CD44. A dibenzocyclooctyne (DBCO)-toxin conjugate, DBCO-MMAE (Monomethylauristatin E), could next target the labeled CSCs in vivo via bioorthogonal Click reaction to achieve excellent anticancer efficacy against a series of tumor models, including orthotopic xenograft, drug-resistant tumor, and lung metastasis with low toxicity. A 5/7 complete remission was observed after single-cycle treatment of an advanced triple-negative breast cancer xenograft (~500 mm3).


Subject(s)
Aldehyde Dehydrogenase , Antibodies , Humans , Azides , Carcinogenesis , Click Chemistry , Aldehyde Dehydrogenase 1 Family , Retinal Dehydrogenase
4.
Biomater Sci ; 9(4): 1301-1312, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33350407

ABSTRACT

The discrepancy of surface receptors on cancerous and non-cancerous cells has been regarded as the mainstay of cancer-targeted therapy. However, due to the heterogeneity of tumor cells and the insufficient levels of receptors on the tumor cell surface, the success of cancer cell-targeted therapies is largely limited. Histone deacetylase/cathepsin l-responsive acetylated azidomannose (DCL-AAM) was previously developed to effectively and selectively label cancer cell surfaces with reactive azido groups via sugar metabolism. Herein, the labeling kinetics and generality of DCL-AAM were systematically investigated in varieties of tumor cells in vitro and in SKOV3 xenograft tumors in vivo. Based on this, dibenzocyclooctyne-cisplatin (DBCO-Pt) prodrug was developed, and DCL-AAM-mediated metabolic labeling of SKOV3 cells enhanced the tumor accumulation of DBCO-Pt ∼2 fold via bioorthogonal click chemistry, potentiating the anti-tumor efficacy of cisplatin yet alleviating the systemic toxicity. This work, therefore, provides the experimental and theoretical support for the future design of sugar metabolism-based targeted delivery systems and may provide a promising candidate for the treatment of cancers lacking appropriate biomarkers.


Subject(s)
Neoplasms , Prodrugs , Cell Line, Tumor , Cisplatin/pharmacology , Click Chemistry , Humans , Neoplasms/drug therapy
5.
Biomater Sci ; 8(19): 5271-5281, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-32744547

ABSTRACT

Glatiramer acetate (GA), a linear random copolypeptide, is a first-line treatment for multiple sclerosis (MS). A major concern, however, is that GA treatment is associated with adverse effects and poor patient adherence due to the need for frequent injections. Here we describe improved performance of the polymeric drug, even at low doses with less-frequent injections, through the modification of its architecture into a star-shaped GA (sGA). In a sGA, multiple GAs are covalently linked onto a core, which greatly changes their properties such as molecular weight, size, and shape. The spherical sGA is retained longer in the body after intraperitoneal injection, and is more readily internalized by RAW 264.7 macrophage cells and bone marrow-derived dendritic cells than GA. In C57BL/6 mice induced with experimental autoimmune encephalitis, a mouse model for MS, sGA treatment exerts disease amelioration effect that is significantly better than that of GA despite a lower dose and less frequent injection. Moreover, spinal cord pathologies of demyelination and leukocyte infiltration are dramatically less pronounced in the sGA treatment condition compared to the GA treatment condition. Thus, we propose that sGA with a higher-ordered architecture offers an attractive and potentially viable treatment option for MS patients.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Glatiramer Acetate , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/drug therapy , Peptides
6.
ACS Omega ; 5(23): 14111-14115, 2020 Jun 16.
Article in English | MEDLINE | ID: mdl-32566878

ABSTRACT

Two Ac4ManNAz (AAM) derivatives with octadecanoic ester (C18 ester) and octadecyl ether (C18 ether) attached to the anomeric hydroxyl groups were synthesized and used in preparation of liposomes. Both liposomes show strong cell-labeling efficiencies on MDA-MB-231 cancer cells. The cell surface-anchored azide group can react with DBCO-Cy5 via Cu-free click chemistry. The two liposomes exhibit different azide placement mechanisms; C18-ether-AAM-treated cells have azido placement through direct insertion, while C18-ester-AAM-treated cells express azido more through metabolic glycoengineering.

7.
Biomaterials ; 238: 119843, 2020 04.
Article in English | MEDLINE | ID: mdl-32062147

ABSTRACT

The early in vivo diagnosis of infectious disease foci is largely hindered by invasion and concealment of pathogens in host cells, making it difficult for conventional probes to detect and analyze intracellular pathogens. Taking advantage of the excessively produced reactive oxygen species (ROS) within host cells, herein we report the design of thiol-hemiketal blocked N-azidoacetyl galactosamine (Ac3GalNAzSP), an azido unnatural sugar bearing an unprecedent designed ROS-responsive moiety for targeted labelling of infected host cells. Ac3GalNAzSP showed great stability under physiological conditions, specifically released active unnatural sugar in host cells overproducing ROS, metabolically labeled infected host cells with azido groups, and enabled targeting in vivo infection sites by subsequent Click Chemistry reactions, substantiating an unprecedented approach for targeting infected host cells. This technique could be a powerful tool for early in vivo diagnosis and targeted treatment of infectious disease.


Subject(s)
Click Chemistry , Sugars , Carbohydrates , Cell Line, Tumor
8.
Bioconjug Chem ; 30(9): 2317-2322, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31403278

ABSTRACT

In comparison with the popular Ac4ManNAz applied as cell labels via Cu-free click chemistry, two novel azido mannosamine lipids with C6 and C12 esters on anomeric hydroxyl groups were prepared and encapsulated in a liposome delivery system, which enhanced chemical stabilities and showed good cell-metabolizable labeling efficiency on MDA-MB-231 cells with strong fluorescence after the treatment of DBCO-Cy5 by triazole formation via click chemistry.


Subject(s)
Azides/chemistry , Hexosamines/chemistry , Liposomes/chemistry , Liposomes/metabolism , Molecular Imaging/methods , Cell Line, Tumor , Click Chemistry , Humans , Staining and Labeling
9.
Biomaterials ; 218: 119305, 2019 10.
Article in English | MEDLINE | ID: mdl-31323538

ABSTRACT

We developed glycopolyesters (GPs) via azido-sugar initiated ring-opening polymerization of O-carboxyanhydrides (OCAs) and achieved efficient in vivo cancer targeting via GP-nanoparticle (GP-NP) mediated metabolic cell labeling followed by Click reaction. GP-NP shows controlled release of azido-sugars and can efficiently label LS174T colon cancer cells with azido groups in tumor-bearing mice. The exogenously introduced azido groups render excellent in vivo cancer targeting and retention of dibenzocyclooctyne-Cy5 (DBCO-Cy5) with an increasing tumor retention enhancement over time (68% at 6 h, 105% at 24 h, and 191% at 48 h) compared to control mice without azido labeling. The tumor accumulation of DBCO-doxorubicin is also significantly enhanced in GP-NP pretreated mice, resulting in improved in vivo anticancer efficacy. This study, for the first time, proposes the use of azido-sugar initiated polymerization of OCAs to form sugar delivery vehicles with high stability and controlled release, and demonstrates the increasing tumor targeting effect of DBCO-cargo over time by azido-modified tumor cells.


Subject(s)
Click Chemistry/methods , Nanoparticles/chemistry , Animals , Cell Line, Tumor , Doxorubicin/chemistry , Drug Design , Mice , Polyesters/chemistry
10.
ACS Appl Mater Interfaces ; 10(46): 39455-39467, 2018 Nov 21.
Article in English | MEDLINE | ID: mdl-30362704

ABSTRACT

Efficient drug accumulation in tumor cells is essential for cancer therapy. Herein, we developed dimeric prodrug self-delivery nanoparticles (NPs) with enhanced drug loading and bioreduction responsiveness for triple negative breast cancer (TNBC) therapy. Specially designed camptothecin dimeric prodrug (CPTD) containing a disulfide bond was constructed to realize intracellular redox potential controlled drug release. Direct conjugation of hydrophobic CPTD to poly(ethylene glycol) PEG5000, a prodrug-based amphiphilic CPTD-PEG5000 co-polymer was synthesized, which could encapsulate parental CPTD prodrug spontaneously and form ultrastable NPs due to the highly analogous structure. Such dimeric prodrug self-delivery nanoparticles showed ultrahigh stability with critical micelle concentration as low as 0.75 µg/mL and remained intact during endocytosis. In addition, neurotensin (NT), a 13 amino acid ligand, was further modified on the nanoparticles for triple negative breast cancer (TNBC) targeting. Optimized NT-CPTD NPs showed improved pharmacokinetics profile and increased drug accumulation in TNBC lesions than free CPT, which largely reduced the systemic toxicity and presented an improved anticancer efficacy in vivo. In summary, with advantages of extremely high drug loading capacity, tumor microenvironmental redox responsiveness, and targeted TNBC accumulation, NT-CPTD NPs showed their potential for effective triple negative breast cancer therapy.


Subject(s)
Drug Carriers/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Apoptosis , Camptothecin/administration & dosage , Cell Line, Tumor , Dimerization , Dithiothreitol/chemistry , Endocytosis , Female , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Ligands , Mice , Micelles , Neurotensin/chemistry , Oxidation-Reduction , Polymers/chemistry , Prodrugs/chemistry , Temperature , Triple Negative Breast Neoplasms/drug therapy
11.
Chem Commun (Camb) ; 54(38): 4878-4881, 2018 May 08.
Article in English | MEDLINE | ID: mdl-29700507

ABSTRACT

In this study, we report incorporation of a covalent linker at the anomeric position of N-azidoacetylmannosamine (ManNAz) for caging its metabolic process. We synthesized a DT-diaphorase-responsive metabolic precursor, HQ-NN-AAM, using an optimized linker. The caged metabolite showed responsiveness to DT-diaphorase in vitro, resulting in metabolic incorporation of an azido sugar into the cell surface in multiple cell lines.


Subject(s)
Azides/metabolism , Cell Tracking/methods , Hexosamines/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Polysaccharides/biosynthesis , Quinones/metabolism , Azides/chemical synthesis , Azides/chemistry , Cell Line, Tumor , Click Chemistry , Ethylenediamines/chemistry , Glycosylation , Hexosamines/chemical synthesis , Hexosamines/chemistry , Humans , Quinones/chemical synthesis , Quinones/chemistry
12.
Chemistry ; 24(29): 7345-7348, 2018 May 23.
Article in English | MEDLINE | ID: mdl-29624762

ABSTRACT

Low cost, high performance hydrolysable polymers are of great importance in biomedical applications and materials industries. While many applications require materials to have a degradation profile insensitive to external pH to achieve consistent release profiles under varying conditions, hydrolysable chemistry techniques developed so far have pH-dependent hydrolytic kinetics. This work reports the design and synthesis of a new type of hydrolysable polymer that has identical hydrolysis kinetics from pH 3 to 11. The unprecedented pH independent hydrolytic kinetics of the aryl ureas were shown to be related to the dynamic bond dissociation controlled hydrolysis mechanism; the resulting hindered poly(aryl urea) can be degraded with a hydrolysis half-life of 10 min in solution. More importantly, these fast degradable hindered aromatic polyureas can be easily prepared by addition polymerization from commercially available monomers and are resistant to hydrolysis in solid form for months under ambient storage conditions. The combined features of good stability in solid state and fast hydrolysis at various pH values is unprecedented in polyurea material, and will have implications for materials design and applications, such as sacrificial coatings and biomaterials.

13.
Biomater Sci ; 6(5): 1189-1200, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29570190

ABSTRACT

Although polymeric nanoconjugates (NCs) hold great promise for the treatment of cancer patients, their clinical utility has been hindered by the lack of efficient delivery of therapeutics to targeted tumor sites. Here, we describe an albumin-functionalized polymeric NC (Alb-NC) capable of crossing the endothelium barrier through a caveolae-mediated transcytosis pathway to better target cancer. The Alb-NC is prepared by nanoprecipitation of doxorubicin (Doxo) conjugates of poly(phenyl O-carboxyanhydrides) bearing aromatic albumin-binding domains followed by subsequent surface decoration of albumin. The administration of Alb-NCs into mice bearing MCF-7 human breast cancer xenografts with limited tumor vascular permeability resulted in markedly increased tumor accumulation and anti-tumor efficacy compared to their conventional counterpart PEGylated NCs (PEG-NCs). The Alb-NC provides a simple, low-cost and broadly applicable strategy to improve the cancer targeting efficiency and therapeutic effectiveness of polymeric nanomedicine.


Subject(s)
Albumins/chemistry , Capillary Permeability , Doxorubicin/chemistry , Endothelium, Vascular/metabolism , Mammary Neoplasms, Experimental/drug therapy , Nanoconjugates/chemistry , Albumins/adverse effects , Albumins/pharmacokinetics , Animals , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Transcytosis
14.
Nano Lett ; 18(3): 1908-1915, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29473753

ABSTRACT

Various delivery vectors have been integrated within biologically derived membrane systems to extend their residential time and reduce their reticuloendothelial system (RES) clearance during systemic circulation. However, rational design is still needed to further improve the in situ penetration efficiency of chemo-drug-loaded membrane delivery-system formulations and their release profiles at the tumor site. Here, a macrophage-membrane-coated nanoparticle is developed for tumor-targeted chemotherapy delivery with a controlled release profile in response to tumor microenvironment stimuli. Upon fulfilling its mission of tumor homing and RES evasion, the macrophage-membrane coating can be shed via morphological changes driven by extracellular microenvironment stimuli. The nanoparticles discharged from the outer membrane coating show penetration efficiency enhanced by their size advantage and surface modifications. After internalization by the tumor cells, the loaded drug is quickly released from the nanoparticles in response to the endosome pH. The designed macrophage-membrane-coated nanoparticle (cskc-PPiP/PTX@Ma) exhibits an enhanced therapeutic effect inherited from both membrane-derived tumor homing and step-by-step controlled drug release. Thus, the combination of a biomimetic cell membrane and a cascade-responsive polymeric nanoparticle embodies an effective drug delivery system tailored to the tumor microenvironment.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Breast Neoplasms/drug therapy , Cell Membrane/metabolism , Delayed-Action Preparations/metabolism , Macrophages/metabolism , Nanoparticles/metabolism , Paclitaxel/administration & dosage , Animals , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Biomimetic Materials/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Delivery Systems , Female , Humans , Mice , Nanoparticles/ultrastructure , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Tumor Microenvironment/drug effects
15.
J Am Chem Soc ; 140(4): 1235-1238, 2018 01 31.
Article in English | MEDLINE | ID: mdl-29332390

ABSTRACT

Polymeric micelles are extensively used for the delivery of hydrophobic drugs, which, however, suffer from unsatisfactory drug loading, colloidal uniformity, formulation stability, and drug release. Herein, we demonstrate a convenient strategy to prepare micelles with ultrahigh drug loading via the incorporation of polymer-drug coordination interactions. An amphiphilic copolymer containing pendant phenylboronic acid as electron acceptor unit was synthesized, which afforded donor-acceptor coordination with doxorubicin to obtain micelles with ultrahigh drug loading (∼50%), nearly quantitative loading efficiency (>95%), uniform size, and colloidal stability. Besides, the encapsulated drug can be effectively and selectively released in response to the high reactive oxygen species levels in cancer cells, which potentiated the anticancer efficacy and reduced systemic toxicity. Apart from doxorubicin, the current platform could be extended to other drugs with electron-donating groups (e.g., epirubicin and irinotecan), rendering a simple and robust strategy for enabling high drug loading in polymeric micelles and cancer-specific drug release.

16.
Nanoscale ; 10(6): 2856-2865, 2018 Feb 08.
Article in English | MEDLINE | ID: mdl-29364314

ABSTRACT

The precise and selective delivery of chemodrugs into tumors represents a critical requirement for anti-cancer therapy. Intelligent delivery systems that are responsive to a single internal or external stimulus often lack sufficient cancer selectivity, which compromises the drug efficacy and induces undesired side effects. To overcome this dilemma, we herein report a cancer-targeting vehicle which allows highly cancer-selective drug release in response to cascaded external (light) and internal (hypoxia) dual triggers. In particular, doxorubicin (DOX)-loaded, hypoxia-dissociable nanoparticles (NPs) were prepared from self-assembled polyethylenimine-nitroimidazole (PEI-NI) micelles that were further co-assembled with hyaluronic acid-Ce6 (HC). Upon accumulation in tumor cells, tumor site-specific light irradiation (660 nm, 10 mW cm-2) generated high levels of reactive oxygen species (ROS) and greatly enhanced the hypoxic levels to induce NP dissociation and accordingly DOX release. A synergistic anti-cancer efficacy between DOX-mediated chemotherapy and Ce6-mediated photodynamic therapy (PDT) was thus achieved, resulting in reduced side effects to normal tissues/cells. This study therefore provides an effective method to control the cancer-specific drug delivery by responding to cascaded multiple triggers, and it renders promising applications for the programmed combination of chemotherapy and PDT toward cancer treatment.


Subject(s)
Doxorubicin/administration & dosage , Drug Delivery Systems , Nanoparticles , Neoplasms, Experimental/therapy , Photochemotherapy , Animals , Cell Line, Tumor , Female , Hyaluronic Acid , Hypoxia , Mice, Inbred C57BL , Micelles , Nitroimidazoles , Photoacoustic Techniques , Polyethyleneimine , Tissue Distribution
17.
Nat Chem Biol ; 13(4): 415-424, 2017 04.
Article in English | MEDLINE | ID: mdl-28192414

ABSTRACT

Distinguishing cancer cells from normal cells through surface receptors is vital for cancer diagnosis and targeted therapy. Metabolic glycoengineering of unnatural sugars provides a powerful tool to manually introduce chemical receptors onto the cell surface; however, cancer-selective labeling still remains a great challenge. Herein we report the design of sugars that can selectively label cancer cells both in vitro and in vivo. Specifically, we inhibit the cell-labeling activity of tetraacetyl-N-azidoacetylmannosamine (Ac4ManAz) by converting its anomeric acetyl group to a caged ether bond that can be selectively cleaved by cancer-overexpressed enzymes and thus enables the overexpression of azido groups on the surface of cancer cells. Histone deacetylase and cathepsin L-responsive acetylated azidomannosamine, one such enzymatically activatable Ac4ManAz analog developed, mediated cancer-selective labeling in vivo, which enhanced tumor accumulation of a dibenzocyclooctyne-doxorubicin conjugate via click chemistry and enabled targeted therapy against LS174T colon cancer, MDA-MB-231 triple-negative breast cancer and 4T1 metastatic breast cancer in mice.


Subject(s)
Breast Neoplasms/metabolism , Carbohydrates/analysis , Carbohydrates/chemistry , Colonic Neoplasms/metabolism , Molecular Probes/analysis , Molecular Probes/metabolism , Molecular Targeted Therapy/methods , Neoplasms, Experimental/metabolism , Animals , Apoptosis/drug effects , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Carbohydrates/chemical synthesis , Cell Line, Tumor , Colonic Neoplasms/diagnosis , Colonic Neoplasms/drug therapy , Dose-Response Relationship, Drug , Drug Design , Drug Screening Assays, Antitumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Molecular Probes/chemical synthesis , Molecular Probes/chemistry , Molecular Structure , Neoplasms, Experimental/diagnosis , Neoplasms, Experimental/drug therapy , Structure-Activity Relationship , Tumor Cells, Cultured
18.
J Control Release ; 263: 211-222, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28062299

ABSTRACT

The past several decades have witnessed the rapid development of nanomedicine (NM) which integrates the advancement of various interdisciplinary areas of science, engineering, and medicine. While a few clinical successes of NM greatly change the landscape of disease diagnosis and treatment, there are several areas of NM remaining to be explored. One such area is the complicated interactions between the NM and biological environment post administration, and how such interaction affects the biological performance of NM. Here, we review the recent progresses on this topic and discuss the interaction of NM with microscopic biomolecules, cells, and the macroscopic in vivo environment. The complete profiling of the bio/nanomaterials interface and interaction should have profound impact on the optimization and de novo design of new NM with better in vivo performance.


Subject(s)
Nanostructures/administration & dosage , Adsorption , Animals , Cells/drug effects , Environment , Humans , Nanostructures/chemistry
19.
Proc Natl Acad Sci U S A ; 113(32): E4601-9, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27457945

ABSTRACT

Malignant osteolysis associated with inoperable primary bone tumors and multifocal skeletal metastases remains a challenging clinical problem in cancer patients. Nanomedicine that is able to target and deliver therapeutic agents to diseased bone sites could potentially provide an effective treatment option for different types of skeletal cancers. Here, we report the development of polylactide nanoparticles (NPs) loaded with doxorubicin (Doxo) and coated with bone-seeking pamidronate (Pam) for the targeted treatment of malignant skeletal tumors. In vivo biodistribution of radiolabeled targeted Pam-NPs demonstrated enhanced bone tumor accumulation and prolonged retention compared with nontargeted NPs. In a murine model of focal malignant osteolysis, Pam-functionalized, Doxo-loaded NPs (Pam-Doxo-NPs) significantly attenuated localized osteosarcoma (OS) progression compared with nontargeted Doxo-NPs. Importantly, we report on the first evaluation to our knowlege of Pam-Doxo-NPs in dogs with OS, which possess tumors of anatomic size and physiology comparable to those in humans. The repeat dosing of Pam-Doxo-NPs in dogs with naturally occurring OS indicated the therapeutic was well tolerated without hematologic, nonhematologic, and cardiac toxicities. By nuclear scintigraphy, the biodistribution of Pam-Doxo-NPs demonstrated malignant bone-targeting capability and exerted measurable anticancer activities as confirmed with percent tumor necrosis histopathology assessment.


Subject(s)
Antineoplastic Agents/administration & dosage , Bone Neoplasms/drug therapy , Diphosphonates/administration & dosage , Doxorubicin/administration & dosage , Nanoconjugates/administration & dosage , Osteolysis/drug therapy , Animals , Diphosphonates/pharmacokinetics , Doxorubicin/toxicity , Female , Mice , Mice, Inbred BALB C , Pamidronate
20.
J Am Chem Soc ; 138(30): 9498-507, 2016 08 03.
Article in English | MEDLINE | ID: mdl-27355522

ABSTRACT

The construction of a multivalent ligand is an effective way to increase affinity and selectivity toward biomolecular targets with multiple-ligand binding sites. Adopting this strategy, we used a known cell-penetrating peptide (CPP) mimic as a scaffold to develop a series of multivalent ligand constructs that bind to the expanded dCTG (CTG(exp)) and rCUG nucleotide repeats (CUG(exp)) known to cause myotonic dystrophy type I (DM1), an incurable neuromuscular disease. By assembling this polyvalent construct, the hydrophobic ligands are solubilized and delivered into cell nuclei, and their enhanced binding affinity leads to the inhibition of ribonuclear foci formation and a reversal of splicing defects, all at low concentrations. Some of the multivalent ligands are shown to inhibit selectively the in vitro transcription of (CTG·CAG)74, to reduce the concentration of the toxic CUG RNA in DM1 model cells, and to show phenotypic improvement in vivo in a Drosophila model of DM1. This strategy may be useful in drug design for other trinucleotide repeat disorders and more broadly for intracellular multivalent targeting.


Subject(s)
Cell-Penetrating Peptides/chemistry , Intracellular Space/metabolism , Peptidomimetics/metabolism , Animals , Animals, Genetically Modified , Biological Transport , Drosophila melanogaster/genetics , HeLa Cells , Humans , Ligands , Models, Molecular , Nucleic Acid Conformation , Peptidomimetics/chemistry , Protein Conformation , Trinucleotide Repeats
SELECTION OF CITATIONS
SEARCH DETAIL
...