Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Article in English | MEDLINE | ID: mdl-38771684

ABSTRACT

Flexible robots (FRs) are generally designed to be lightweight to achieve rapid motion. However, accompanying vibrations and modeling errors influence tracking control, especially in situations involving reference signal loss. This article develops a two-time scale primal-dual inverse reinforcement learning (PD-IRL) framework for FRs to perform tracking tasks with incomplete reference signals. First, consider the admissible policy as a nonconvex input constraint to guarantee the stable operation of the equipment. Then, FRs imitate the demonstration behaviors of an expert, including both rigid and flexible motions, to achieve a balance in tracking speed and vibration suppression. During the imitation process, nonconvex optimization problems of FRs are transformed into corresponding dual problems to obtain the global optimal policy. Moreover, employing multiple linearly independent paths to explore the state space simultaneously can improve convergence speed. Convergence and stability are studied rigorously. Finally, simulations and comparisons show the effectiveness and superiority of the proposed method.

2.
J Mater Chem B ; 11(16): 3679-3692, 2023 04 26.
Article in English | MEDLINE | ID: mdl-37042187

ABSTRACT

Although oxidative stress-based antitumor modality derived from reactive oxygen species (ROS) storm has attracted considerable attention in copper-based nanomaterials, its efficiency is still weakened by the insufficient hydrogen peroxide (H2O2) and overexpressed glutathione (GSH) in a tumor microenvironment (TME). In view of this, we designed an engineered programmable spike-like nanogenerator via the coordination-driven co-assembly of Evans Blue (EB), copper ions (CuII), and 5-hydroxy-p-naphthoquinone (HND). For programmable nanogenerators, the introduction of EB as a stabilizer-like component can not only adjust its morphology but also achieve its visual tracking. Interestingly, such programmable nanogenerators can be efficiently enriched in tumor regions and then internalized into tumor cells due to ECH with spike-like morphology. Notably, once the nanogenerator is disintegrated and burst to release the drug upon acidic lysosome and endogenous GSH triggering, the released HND can not only efficiently amplify endogenous H2O2 by intracellular oxidoreductases but also down-regulate the peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin 1) activity. In addition, the released CuII ions can efficiently catalyze the degradation of the endogenous H2O2 to amplify hydroxyl radicals (˙OH) and down-regulate the overexpressed GSH to reduce ˙OH elimination for on-demand cascade-amplifying oxidative stress. Importantly, such programmable nanogenerators show an excellent antitumor effect via down-regulating the Pin 1 activity and cascade-amplifying oxidative stress. In this study, we propose a spatiotemporally programmable cascade nanogenerator for oxidative stress-based antitumor therapy.


Subject(s)
Copper , Hydrogen Peroxide , Copper/pharmacology , Hydrogen Peroxide/metabolism , Tumor Microenvironment , Oxidative Stress , Reactive Oxygen Species/metabolism , Glutathione/metabolism
3.
ACS Appl Mater Interfaces ; 15(14): 17495-17506, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-36996342

ABSTRACT

How to efficiently synthesize toxic chemo-drugs in the hypoxia tumor microenvironment still faces a huge challenge. Herein, we have tailored engineered vehicle-free nanoreactors by coordination-driven co-assembly of photosensitizer indocyanine green (ICG), transition metal platinum (Pt), and nontoxic 1,5-dihydroxynaphthalene (DHN) to self-amplify O2 and cascade chemo-drug synthesis in tumor cells for self-reinforcing hypoxic oncotherapy. Once vehicle-free nanoreactors are internalized into tumor cells, they show a serious instability that results in rapid disassembly and on-demand drug release under the stimuli of acidic lysosome and laser radiation. Notably, the released Pt can efficiently decompose the endogenous hydrogen peroxide (H2O2) into O2 to alleviate tumor hypoxia, which is conducive to enhancing the photodynamic therapy (PDT) efficiency of the released ICG. Complementarily, a large amount of the 1O2 generated by PDT can efficiently oxidize the released nontoxic DHN into the highly toxic chemo-drug juglone. Therefore, such vehicle-free nanoreactors can achieve intracellular on-demand cascade chemo-drug synthesis and self-reinforce photo-chemotherapeutic efficacy on the hypoxic tumor. On the whole, such a simple, flexible, efficient, and nontoxic therapeutic strategy will broaden the study of on-demand chemo-drug synthesis and hypoxic oncotherapy.


Subject(s)
Antineoplastic Agents , Nanoparticles , Neoplasms , Photochemotherapy , Humans , Photochemotherapy/methods , Platinum/therapeutic use , Hydrogen Peroxide , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Hypoxia/drug therapy , Nanotechnology , Cell Line, Tumor , Tumor Microenvironment
4.
ACS Appl Mater Interfaces ; 14(4): 5033-5052, 2022 Feb 02.
Article in English | MEDLINE | ID: mdl-35045703

ABSTRACT

Although nanotheranostics have displayed striking potential toward precise nanomedicine, their targeting delivery and tumor penetration capacities are still impeded by several biological barriers. Besides, the current antitumor strategies mainly focus on killing tumor cells rather than antiangiogenesis. Enlightened by the fact that the smart transformable self-targeting nanotheranostics can enhance their targeting efficiency, tumor penetration, and cellular uptake, we herein report carrier-free Trojan-horse diameter-reducible metal-organic nanotheranostics by the coordination-driven supramolecular sequential co-assembly of the chemo-drug pemetrexed (PEM), transition-metal ions (FeIII), and antiangiogenesis pseudolaric acid B. Such nanotheranostics with both a high dual-drug payload efficiency and outstanding physiological stability are responsively decomposed into numerous ultra-small-diameter nanotheranostics under stimuli of the moderate acidic tumor microenvironment and then internalized into tumor cells through tumor-receptor-mediated self-targeting, synergistically enhancing tumor penetration and cellular uptake. Besides, such nanotheranostics enable visualization of self-targeting capacity under the macroscopic monitor of computed tomography/magnetic resonance imaging, thereby realizing efficient oncotherapy. Moreover, tumor microvessels are precisely monitored by optical coherence tomography angiography/laser speckle imaging during chemo-antiangiogenic therapy in vivo, visually verifying that such nanotheranostics possess an excellent antiangiogenic effect. Our work will provide a promising strategy for further tumor diagnosis and targeted therapy.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Biocompatible Materials/pharmacology , Metal-Organic Frameworks/pharmacology , Neovascularization, Pathologic/drug therapy , Theranostic Nanomedicine , Angiogenesis Inhibitors/chemistry , Antineoplastic Agents/chemistry , Biocompatible Materials/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/chemistry , Diterpenes/pharmacology , Drug Screening Assays, Antitumor , Humans , Materials Testing , Metal-Organic Frameworks/chemistry , Neovascularization, Pathologic/pathology , Particle Size , Pemetrexed/chemistry , Pemetrexed/pharmacology , Surface Properties
5.
J Colloid Interface Sci ; 603: 70-84, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34186412

ABSTRACT

Nanotherapy based on thermochemotherapy has boomed as a promising alternative for oncotherapy due to the enhanced permeability and retention (EPR) effect. However, a lack of self-targeting capacity prevents nanotherapy from efficiently accumulating in tumor tissue and internalizing into tumor cells, resulting in a suboptimal therapeutic effect. To overcome these bottlenecks, a kind of methotrexate (MTX)-soybean phospholipid (SPC) inclusion complex (MTX-SPC)-modified graphene oxide (CGO) nanotherapy (CGO-MTX-SPC) is constructed by CGO nanosheets as a supporter for MTX-SPC, thereby realizing active-targeting and synergistic thermochemotherapy. As an FDA-approved chemotherapeutic drug, MTX can be regarded as a tumor-targeting enhancer against the folate receptor on account of its similar structure to folic acid (FA). The fabricated CGO-MTX-SPC has a sheet shape with a size of ca. 109 nm and tumor microenvironment-responsive on-demand drug release. It is worth noting that the physiological stability of CGO-MTX-SPC is better than that of CGO while displaying an improved photothermal effect. In addition, CGO-MTX-SPC can specifically recognize tumor cells and then achieve on-demand drug burst release by dual stimuli of internal lysosomal acidity and an external laser. Moreover, in vivo experimental results further demonstrate that CGO-MTX-SPC displays significant enrichment at the tumor location by active targeting mechanisms due to the introduction of MTX-SPC, endowing the synergistic thermochemotherapy effect upon 808 nm laser irradiation and almost thorough tumor elimination while significantly erasing undesirable side effects. Taken together, the design idea of our nanotherapy not only provides a potential tumor-targeting therapeutic strategy but also broadens the drug payload method of two-dimensional nanomaterials.


Subject(s)
Drug Delivery Systems , Nanoparticles , Cell Line, Tumor , Cell Survival , Graphite , Methotrexate
6.
J Mater Chem B ; 9(27): 5547-5559, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34165487

ABSTRACT

Sonodynamic therapy has attracted wide attention as a noninvasive therapy due to deep tissue penetration. However, majority sonosensitizers often suffer from poor physiological stability, rapid blood clearance and nonspecific targeting, which seriously hinders their further practical applications. Inspired by the concept of active targeting drug delivery, both dual-functional chemo-drug pemetrexed (PEM, emerges an innate affinity toward the folate receptor) and amphiphilic d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) were selected to be covalently linked by an esterase-responsive ester linkage. The synthesized self-targeting TPGS-PEM prodrug and indocyanine green (ICG) as functional motifs can be self-assembled into a TPGS-PEM-ICG nanoplatform within an aqueous medium. The TPGS-PEM-ICG nanoplatform with outstanding structural and physiological stability not only protects the sonosensitizer from reticular endothelial system clearance but also achieves active targeting drug delivery and efficient tumor enrichment. Moreover, TPGS-PEM-ICG nanoplatform can selectively recognize tumor cells and then realize on-demand drug burst release by multiple stimuli of internal lysosomal acidity, esterase and external ultrasound, which guarantee low side effects toward normal tissues and organs. It is also worth noting that our nanoplatform exhibits protruding tumor enrichment under the precise guidance of photoacoustic/fluorescence imaging. Further in vitro and in vivo experimental results well confirmed that the TPGS-PEM-ICG nanoplatform possesses enhanced chemo-sonodynamic effects. Interestingly, the highly toxic reactive oxygen species can remarkably reduce the blood oxygen saturation signal of the tumor microenvironment via precise, multifunctional and high-resolution photoacoustic imaging. Taken together, the TPGS-PEM-ICG nanoplatform can be expected to hold enormous potential for diagnosis, prognosis and targeted therapy for tumor.


Subject(s)
Antineoplastic Agents/pharmacology , Indocyanine Green/chemistry , Nanoparticles/chemistry , Photoacoustic Techniques , Prodrugs/pharmacology , Ultrasonic Therapy , Vitamin E/pharmacology , Animals , Antineoplastic Agents/chemistry , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Humans , Mice , Molecular Structure , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/drug therapy , Optical Imaging , Particle Size , Prodrugs/chemistry , Vitamin E/chemistry
7.
Int J Med Sci ; 18(9): 1999-2007, 2021.
Article in English | MEDLINE | ID: mdl-33850470

ABSTRACT

Precartilaginous stem cells (PCSCs) are able to initiate chondrocyte and bone development. The present study aimed to investigate the role of miR-143 and the underlying mechanisms involved in PCSC proliferation. In a rat growth plate injury model, tissue from the injury site was collected and the expression of miR-143 and its potential targets was determined. PCSCs were isolated from the rabbits' distal epiphyseal growth plate. Cell viability, DNA synthesis, and apoptosis were determined with MTT, BrdU, and flow cytometric analysis, respectively. Real time PCR and western blot were performed to detect the mRNA and protein expression of the indicated genes. Indian hedgehog (IHH) was identified as a target gene for miR-143 with luciferase reporter assay. Decreased expression of miR-143 and increased expression of IHH gene were observed in the growth plate after injury. miR-143 mimics decreased cell viability and DNA synthesis and promoted apoptosis of PCSCs. Conversely, siRNA-mediated inhibition of miR-143 led to increased growth and suppressed apoptosis of PCSCs. Transfection of miR-143 decreased luciferase activity of wild-type IHH but had no effect when the 3'-UTR of IHH was mutated. Furthermore, the effect of miR-143 overexpression was neutralized by overexpression of IHH. Our study showed that miR-143 is involved in growth plate behavior and regulates PCSC growth by targeting IHH, suggesting that miR-143 may serve as a novel target for PCSC-related diseases.


Subject(s)
Growth Plate/pathology , Hedgehog Proteins/genetics , MicroRNAs/metabolism , Salter-Harris Fractures/pathology , Stem Cells/metabolism , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cells, Cultured , Disease Models, Animal , Growth Plate/cytology , Growth Plate/growth & development , Humans , Primary Cell Culture , Rabbits , Rats , Salter-Harris Fractures/therapy , Stem Cell Transplantation
8.
Int J Pharm ; 594: 120184, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33340597

ABSTRACT

D-a-tocopheryl polyethylene glycol succinate (TPGS) as a FDA-approved safe adjuvant has shown an excellent application in the targeting delivery of antitumor drugs and overcoming multidrug resistance. Beside, TPGS can result in apoptogenic activity toward many tumor types because it can induce mitochondrial dysfunction. Therefore, TPGS can serve as an antineoplastic agent. However, the current research on the selective antitumor activity of TPGS is ignored. To reveal the issue, herein we develop a mitochondria-targeting drug-free TPGS nanomicelles with the hydrodynamic diameter of about 100 nm and outstanding serum stability by weak interaction-driven self-assembly of the amphiphilic TPGS polymer. Moreover, such drug-free TPGS nanomicelles intravenously injected into tumor-bearing mice exhibit long blood circulation time, superior tumor enrichment, and inhibit the tumor growth via inducing excessive reactive oxygen species (ROS) generation within tumor cells. Further in vitro and in vivo researches jointly demonstrate that drug-free TPGS nanomicelles have more significant antitumor effect on HeLa cells compared with that of other tumor cells. On the contrary, drug-free TPGS nanomicelles display the low toxicity toward normal cells and tissues. Taken together, these new findings confirm that TPGS drug-free nanomicelles represent simple, multifunctional, safe, and efficient antineoplastic agents, which can be expected to bring new light on the development of drug-free polymers for tumor therapy.


Subject(s)
Antineoplastic Agents , Polyethylene Glycols , Animals , Antineoplastic Agents/pharmacology , Cell Death , Cell Line, Tumor , HeLa Cells , Humans , Mice , Micelles , Mitochondria , Reactive Oxygen Species , Vitamin E
9.
Mol Pharm ; 17(7): 2435-2450, 2020 07 06.
Article in English | MEDLINE | ID: mdl-32459486

ABSTRACT

On-demand drug release nanoplatforms are promising alternative strategies for enhancing the therapeutic effect of cancer chemotherapy. However, these nanoplatforms still have many drawbacks including rapid blood clearance, nontargeted specificity, and a lack of immune escape function. Even worse, they are also hindered via the dosage-limiting toxicity of traditional chemotherapeutic drugs. Herein, both dual-functional mannose (enhances the antitumor activity of chemotherapeutic drugs and exhibits an innate affinity against the lectin receptor) and amphiphilic d-α-tocopheryl polyethylene glycol 1000 succinate were selected to be covalently linked via a redox-responsive monothioether linkage. The synthesized self-distinguished polymer (TSM), as a structural motif, can be self-assembled into nanoparticles (TSM NPs) in an aqueous solution, in which doxorubicin (DOX) is loaded by weak interactions (TSM-DOX NPs). These TSM-DOX NPs can provide targeted, on-demand drug release under dual stimuli from lysosomal acidity and glutathione (GSH). In addition, TSM-DOX NPs can be self-distinguished via tumor cells in vitro and specifically self-distinguished from the tumor site in vivo. Further in vitro and in vivo research consistently demonstrated that TSM-DOX NPs display highly synergistic chemotherapeutic effects. Taken together, the data show that the self-distinguished GSH-responsive polymer TSM has the potential to load various therapeutic agents.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Drug Liberation , Nanoparticles/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Animals , Apoptosis/drug effects , Cell Survival/drug effects , Female , Hep G2 Cells , Humans , MCF-7 Cells , Male , Mice , Mice, Inbred BALB C , Mice, Nude , NIH 3T3 Cells , Rats , Rats, Sprague-Dawley , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
10.
Biosci Rep ; 40(5)2020 05 29.
Article in English | MEDLINE | ID: mdl-32319512

ABSTRACT

Precartilaginous stem cells (PCSCs) are adult stem cells that can initiate chondrocytes and bone development. In the present study, we explored whether miR-132/212 was involved in the proliferation of PCSCs via Hedgehog signaling pathway. PCSCs were isolated and purified with the fibroblast growth factor receptor-3 (FGFR-3) antibody. Cell viability, DNA synthesis and apoptosis were measured using MTT, BrdU and flow cytometric analysis. The mRNA and protein expression were detected by real-time PCR and Western blot, respectively. The target gene for miR-132/212 was validated by luciferase reporter assay. Results showed that transfection with miR-132/212 mimic significantly increased cell viability and DNA synthesis, and inhibited apoptosis of PCSCs. By contrast, miR-132/212 inhibitor could suppress growth and promote apoptosis of PCSCs. Luciferase reporter assays indicated that transfection of miR-132/212 led to a marked reduction of luciferase activity, but had no effect on PTCH1 3'-UTR mutated fragment, suggesting that Patched1 (PTCH1) is a target of miR-132/212. Furthermore, treatment with miR-132/212 mimics obviously increased the protein expression of Indian hedgehog (Ihh) and parathyroid hormone related protein (PTHrP), which was decreased after treatment with Hedgehog signaling inhibitor, cyclopamine. We also found that inhibition of Ihh/PTHrP signaling by cyclopamine significantly suppressed growth and DNA synthesis, and induced apoptosis in PCSCs. These findings demonstrate that miR-132/212 promotes growth and inhibits apoptosis in PCSCs by regulating PTCH1-mediated Ihh/PTHrP pathway, suggesting that miR-132/212 cluster might serve as a novel target for bone diseases.


Subject(s)
Adult Stem Cells/physiology , Cell Proliferation/genetics , Chondrocytes/physiology , MicroRNAs/metabolism , Animals , Apoptosis/drug effects , Cartilage, Articular/cytology , Cell Proliferation/drug effects , Cells, Cultured , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Multigene Family , Parathyroid Hormone-Related Protein/metabolism , Patched-1 Receptor/metabolism , Primary Cell Culture , Rabbits , Veratrum Alkaloids/pharmacology
11.
ACS Appl Mater Interfaces ; 12(13): 14884-14904, 2020 Apr 01.
Article in English | MEDLINE | ID: mdl-32167740

ABSTRACT

Low drug payload and lack of tumor-targeting for chemodynamic therapy (CDT) result in an insufficient reactive oxygen species (ROS) generation, which seriously hinders its further clinical application. Therefore, how to improve the drug payload and tumor targeting for amplification of ROS and combine it with chemotherapy has been a huge challenge in CDT. Herein, methotrexate (MTX), gadolinium (Gd), and artesunate (ASA) were used as theranostic building blocks to be coordinately assembled into tumor-specific endogenous FeII-activated and magnetic resonance imaging (MRI)-guided self-targeting carrier-free nanoplatforms (NPs) for amplification of ROS and enhanced chemodynamic chemotherapy. The obtained ASA-MTX-GdIII NPs exhibited extremely high drug payload (∼96 wt %), excellent physiological stability, long circulating ability (half-time: ∼12 h), and outstanding tumor accumulation. Moreover, ASA-MTX-GdIII NPs could be specifically uptaken by tumor cells via folate (FA) receptors and subsequently be disassembled via lysosomal acidity-induced coordination breakage, resulting in drug burst release. Most strikingly, the produced ASA could be catalyzed by tumor-specific overexpressed endogenous FeII ions to generate sufficient ROS for enhancing the main chemodynamic efficacy, which could exert a synergistic effect with the assistant chemotherapy of MTX. Interestingly, ASA-MTX-GdIII NPs caused a lower ROS generation and toxicity on normal cell lines that seldom expressed endogenous FeII ions. Under MRI guidance with assistance of self-targeting, significantly superior synergistic tumor therapy was performed on FA receptor-overexpressed tumor-bearing mice with a higher ROS generation and an almost complete elimination of tumor. This work highlights ASA-MTX-GdIII NPs as an efficient chemodynamic-chemotherapeutic agent for MRI imaging and tumor theranostics.


Subject(s)
Antineoplastic Agents/chemistry , Ferrous Compounds/chemistry , Gadolinium/chemistry , Nanoparticles/chemistry , Theranostic Nanomedicine , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Artesunate/chemistry , Artesunate/pharmacology , Artesunate/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Drug Carriers/chemistry , Humans , Hydrogen-Ion Concentration , Magnetic Resonance Imaging , Methotrexate/chemistry , Methotrexate/pharmacology , Methotrexate/therapeutic use , Mice , Mice, Nude , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Neoplasms/pathology , Rats , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
12.
ACS Appl Mater Interfaces ; 11(17): 15262-15275, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30964624

ABSTRACT

Nanoparticles camouflaged by red blood cell (RBC) membranes have attracted considerable attention owing to reservation of structure of membrane and surface proteins, endowing prominent cell-specific function including biocompatibility, prolonged circulation lifetime, and reduced reticular endothelial system (RES) uptake ability. Considering the drawbacks of carrier-free nanomedicine including the serious drug burst release, poor stability, and lack of immune escape function, herein we developed and fabricated a novel RBC membranes biomimetic combinational therapeutic system by enveloping the small molecular drug coassemblies of 10-hydroxycamptothecin (10-HCPT) and indocyanine green (ICG) in the RBC membranes for prolonged circulation, controlled drug release, and synergistic chemo-photothermal therapy (PTT). The self-reorganized RBCs@ICG-HCPT nanoparticles (NPs) exhibited a diameter of ∼150 nm with core-shell structure, high drug payload (∼92 wt %), and reduced RES uptake function. Taking advantage of the stealth functionality of RBC membranes, RBCs@ICG-HCPT NPs remarkably enhanced the accumulation at the tumor sites by passive targeting followed by cellular endocytosis. Upon the stimuli of near-infrared laser followed by acidic stimulation, RBCs@ICG-HCPT NPs showed exceptional instability by heat-mediated membrane disruption and pH change, thereby triggering the rapid disassembly and accelerated drug release. Consequently, compared with individual treatment, RBCs@ICG-HCPT NPs under dual-stimuli accomplished highly efficient apoptosis in cancer cells and remarkable ablation of tumors by chemo-PTT. This biomimetic nanoplatform based on carrier-free, small molecular drug coassemblies integrating imaging capacity as a promising theranostic system provides potential for cancer diagnosis and combinational therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/chemistry , Biomimetics , Camptothecin/analogs & derivatives , Cell Membrane/chemistry , Infrared Rays , Nanoparticles/chemistry , Neoplasms/therapy , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Camptothecin/chemistry , Camptothecin/pharmacology , Camptothecin/therapeutic use , Erythrocytes/cytology , Erythrocytes/metabolism , HeLa Cells , Humans , Hydrogen-Ion Concentration , Indocyanine Green/chemistry , Male , Mice , Mice, Nude , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Phototherapy , Rats , Rats, Sprague-Dawley
13.
Genomics ; 111(3): 320-326, 2019 05.
Article in English | MEDLINE | ID: mdl-29486210

ABSTRACT

Developmental dysplasia of the hip (DDH) is a common congenital malformation characterized by mismatch in shape between the femoral head and acetabulum, and leads to hip dysplasia. To date, the pathogenesis of DDH is poorly understood and may involve multiple factors, including genetic predisposition. However, comprehensive genetic analysis has not been applied to investigate a genetic component of DDH. In the present study, 10 pairs of healthy fathers and DDH daughters were enrolled to identify genetic hallmarks of DDH using high throughput whole genome sequencing. The DDH-specific DNA mutations were found in each patient. Overall 1344 genes contained DDH-specific mutations. Functional enrichment analysis showed that these genes played important roles in the cytoskeleton, microtubule cytoskeleton, sarcoplasm and microtubule associated complex. These functions affected osteoblast and osteoclast development. Therefore, we proposed that the DDH-specific mutations might affect bone development, and caused DDH. Our pairwise high throughput sequencing results comprehensively delineated genetic hallmarks of DDH. Further research into the biological impact of these mutations may inform the development of DDH diagnostic tools and allow neonatal gene screening.


Subject(s)
Hip Dislocation, Congenital/genetics , Mutation , Adult , Child, Preschool , Female , Hip Dislocation, Congenital/pathology , Humans , Infant , Male , Osteogenesis/genetics , Pedigree , Whole Genome Sequencing
14.
ACS Biomater Sci Eng ; 5(9): 4442-4454, 2019 Sep 09.
Article in English | MEDLINE | ID: mdl-33438410

ABSTRACT

The medical application of nanotechnology is promising for cancer chemotherapy. However, most of the small-molecule drug assemblies still have such disadvantages as serious drug leakage and nonideal synergistic mechanisms, resulting in undesired therapeutic effect. Both nucleoside analogue-based clofarabine (CA) and methotrexate (MTX) were used as the first-line anticancer medication. However, a single-agent chemotherapy still faced many challenges including low bioavailability and toxic side effects to normal tissues due to nonspecific biodistribution of drugs. Herein, we designed and fabricated novel viral-mimicking and carry-free nanodrugs (CA-MTX NPs) via molecular recognition-driven precise self-assembly between CA and MTX. After introduction of mild acid-responsive PEG-lipid on the surface of CA-MTX NPs, the synthetic nanodrugs with a diameter of ∼150 nm exhibited tumor microenvironment-activated characteristics and self-targeting property. The results suggested that our nanodrugs could achieve superior tumor accumulation and synergistically promote the tumor suppression by collaboratively inhibiting dNTP pools. We foresaw that the well-designed smart nanodrugs delivery system would open a new avenue in synergistic cancer therapeutics.

15.
Int J Pharm ; 549(1-2): 230-238, 2018 Oct 05.
Article in English | MEDLINE | ID: mdl-30071310

ABSTRACT

Poor water solubility, short half-life, and low drug efficacy posed a challenge for clinical application of curcumin (CUR). In this work, a kind of CUR prodrug was synthesized by coupling two CUR molecules with a mono-thioether linker for glutathione (GSH)-responsive drug delivery. The synthesized CUR prodrug (CUR-S-CUR dimer) could self-assemble into the homogeneous spherical nanoparticles (NPs) in aqueous solution followed by surface functionalization of trace amounts of DSPE-PEG. These CUR-S-CUR@PEG NPs exhibited a small particle size of ∼100 nm, high CUR-loading content of ∼78 wt%, and good colloid stability. Moreover, the CUR-S-CUR@PEG NPs demonstrated much more efficient cellular uptake and intracellular/nuclear drug delivery compared with free CUR, indicating the advantages of small molecular prodrug assembly. In addition, the GSH with high concentration in tumor cells could trigger the disassembly of CUR-S-CUR@PEG NPs. Furthermore, the cytotoxicity assays indicated that the CUR-S-CUR@PEG NPs exhibited the comparable inhibition effect of tumor cell proliferation with free CUR due to sustained drug release. Therefore, these stimuli-responsive CUR-S-CUR@PEG NPs might have promising potential for highly efficient intracellular drug delivery and controlled drug release in cancer therapy.


Subject(s)
Curcumin/administration & dosage , Drug Delivery Systems , Glutathione/metabolism , Nanoparticles , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Chemistry, Pharmaceutical/methods , Curcumin/pharmacokinetics , Curcumin/pharmacology , Delayed-Action Preparations , Drug Carriers/chemistry , Drug Stability , Half-Life , HeLa Cells , Humans , Particle Size , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Prodrugs , Solubility
16.
Biomater Sci ; 6(7): 1818-1833, 2018 Jun 25.
Article in English | MEDLINE | ID: mdl-29785434

ABSTRACT

Combinational cancer therapy has been considered as a promising strategy to achieve synergetic therapeutic effects and suppression of multidrug resistance. Herein, we adopted a combination of methotrexate (MTX), an antimetabolite acting on cytoplasm, and 10-hydroxycamptothecin (HCPT), an alkaloid acting on nuclei, to treat cancer. Given the different solubilities, membrane permeabilities, and anticancer mechanisms of both drugs, we developed a dual-targeting delivery system based on 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-hyaluronic acid (a principal ligand of CD44 receptors)-MTX (a selective ligand of folate receptors) nanoparticles, which was exploited to carry HCPT-MTX conjugate for synergistically boosting dual-drug co-delivery. The HCPT-MTX conjugate was synthesized by a blood-stable yet intracellularly hydrolysable ester bond. The core-shell-corona DSPE-HA-MTX nanoparticles encapsulating HCPT-MTX (HCPT-MTX@DHM) exhibited high drug entrapment efficiency (∼91.8%) and pH/esterase-controlled release behavior. Cellular uptake studies confirmed significant increase in the efficiency of selective internalization of HCPT-MTX@DHM via CD44/folate receptors compared with those of DSPE-HA nanoparticles encapsulating HCPT-MTX (HCPT-MTX@DH), both drugs, or each individual drug. Furthermore, in vivo near-infrared fluorescence and photoacoustic dual-modal imaging indicated that DiR-doped HCPT-MTX@DHM nanoparticles efficiently accumulated at the tumor sites through passive-plus-active targeting. Finally, the synergistic active targeting and synchronous dual-drug release at a synergistic drug-to-drug ratio resulted in highly synergetic tumor cell-killing and tumor growth inhibition in MCF-7 tumor-bearing mice. Therefore, HCPT-MTX@DHM nanoparticles can be an efficient and smart platform for tumor-targeting therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Camptothecin/analogs & derivatives , Hyaluronic Acid/chemistry , Methotrexate/pharmacology , Phosphatidylethanolamines/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Camptothecin/chemistry , Camptothecin/pharmacology , Drug Carriers , Drug Compounding/methods , Drug Synergism , Female , Folate Receptors, GPI-Anchored/genetics , Folate Receptors, GPI-Anchored/metabolism , Folic Acid/metabolism , Humans , MCF-7 Cells , Methotrexate/chemistry , Mice , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Nanoparticles/metabolism , Survival Analysis , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
17.
Int J Nanomedicine ; 13: 1381-1398, 2018.
Article in English | MEDLINE | ID: mdl-29563794

ABSTRACT

AIM: We designed acid-labile methotrexate (MTX) targeting prodrug self-assembling nanoparticles loaded with curcumin (CUR) drug for simultaneous delivery of multi-chemotherapeutic drugs and combination cancer therapy. METHODS: A dual-acting MTX, acting as both an anticancer drug and as a tumor-targeting ligand, was coupled to 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[aldehyde(polyethylene glycol)-2000] via Schiff's base reaction. The synthesized prodrug conjugate (DSPE-PEG-Imine-MTX) could be self-assembled into micellar nanoparticles (MTX-Imine-M) in aqueous solution, which encapsulated CUR into their core by hydrophobic interactions (MTX-Imine-M-CUR). RESULTS: The prepared MTX-Imine-M-CUR nanoparticles were composed of an inner hydrophobic DSPE/CUR core and an outside hydrophilic bishydroxyl poly (ethyleneglycol) (PEG) shell with a self-targeting MTX prodrug corona. The imine linker between 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[aldehyde(polyethyleneglycol)-2000] and MTX, as a dynamic covalent bond, was strong enough to remain intact in physiological pH, even though it is rapidly cleaved in acidic pH. The MTX-Imine-M-CUR could codeliver MTX and CUR selectively and efficiently into the cancer cells via folate receptor-mediated endocytosis followed by the rapid intracellular release of CUR and the active form of MTX via the acidity of endosomes/lysosomes. Moreover, the MTX-Imine-M-CUR resulted in significantly higher in vitro and in vivo anticancer activity than pH-insensitive DSPE-PEGAmide-MTX assembling nanoparticles loaded with CUR (MTX-Amide-M-CUR), MTX unconjugated DSPE-PEG assembling micellar nanoparticles loaded with CUR (M-CUR), combination of both free drugs, and individual free drugs. CONCLUSION: The smart system provided a simple, yet feasible, drug delivery strategy for targeted combination chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Curcumin/therapeutic use , Drug Delivery Systems/methods , Methotrexate/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Death/drug effects , Colloids , Curcumin/chemistry , Curcumin/pharmacology , Drug Carriers/chemistry , Drug Liberation , Endocytosis/drug effects , HeLa Cells , Humans , Hydrodynamics , Hydrogen-Ion Concentration , MCF-7 Cells , Methotrexate/pharmacology , Mice, Inbred BALB C , Mice, Nude , Micelles , Particle Size , Phosphatidylethanolamines/chemical synthesis , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Prodrugs/pharmacology
18.
ACS Appl Mater Interfaces ; 9(45): 39127-39142, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-29039650

ABSTRACT

Tumor-targeting combination chemotherapy is an important way to improve the therapeutic index and reduce the side effects as compared to traditional cancer treatments. However, one of the major challenges in surface functionalization of nanoparticle (NP) is accomplishing multiple purposes through one single ligand. Upon such consideration, methotrexate (MTX), an anticancer drug with a targeting moiety inspired by the similar structure of folate, could be used to covalently link with lipid-polymer conjugate (DSPE-PEG) via a pH-sensitive dynamic covalent imine (CH═N) bond to synthesize the acid-induced function "targeting-anticancer" switching DSPE-PEG-CH═N-MTX. We hypothesize that using this kind of MTX prodrug to functionalize NP's surface would be conductive to combine the early phase active targeting function and the late-phase anticancer function in one nanosystem. Herein, a nanococktail is programmed for codelivery of epirubicin (EPI) and MTX by co-self-assembly of acid-dissociated EPI-phospholipid (PC) complex and acid-cleavable DSPE-PEG-CH═N-MTX conjugate. The obtained nanococktail (MTX-PEG-EPI-PC NPs) could not only actively target folate receptors-overexpressing tumor cells but also respond to acidic endo/lysosomes for triggering the on-demand release of pharmaceutically active EPI/MTX. The intracellular drug distribution also demonstrated that the system could codeliver two drugs to individual target sites of action, inducing the significant synergistic anticancer efficiency based on different anticancer mechanisms. More importantly, the in vivo tumor accumulation and anticancer efficacy of MTX-PEG-EPI-PC NPs (via cleavable imine bond) were significantly enhanced as compared to the individual free drug, both free drugs, PEG-EPI-PC NPs, and MTX-PEG-EPI-PC NPs (via the uncleavable amide bond). This self-synergistic tumor-targeting therapy might represent a promising strategy for cancer treatment.


Subject(s)
Neoplasms , Antineoplastic Agents , Drug Delivery Systems , Humans , Hydrogen-Ion Concentration , Methotrexate , Nanoparticles , Polyethylene Glycols , Prodrugs
19.
Colloids Surf B Biointerfaces ; 160: 649-660, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-29031225

ABSTRACT

As one of nanomedicine delivery systems (NDSs), drug nanocrystals exhibited an excellent anticancer effect. Recently, differences of internalization mechanisms and subcellular localization of both drug nanocrystals and small molecular free drug have drawn much attention. In this paper, paclitaxel (PTX) as a model anticancer drug was directly labeled with 4-chloro-7-nitro-1, 2, 3-benzoxadiazole (NBD-Cl) (a drug-fluorophore conjugate Ma et al. (2016) and Wang et al. (2016) [1,2] (PTX-NBD)). PTX-NBD was synthesized by nucleophilic substitution reaction of PTX with NBD-Cl in high yield and characterized by fluorescence, XRD, ESI-MS, and FT-IR analysis. Subsequently, the cube-shaped PTX-NBD nanocrystals were prepared with an antisolvent method followed by surface functionalization of SPC and MPEG-DSPE. The obtained specific shaped PTX-NBD@PC-PEG NCs had a hydrodynamic particle size of ∼50nm, excellent colloidal stability, and a high drug-loading content of ∼64%. Moreover, in comparison with free PTX-NBD and the sphere-shaped PTX-NBD nanocrystals with surface functionalization of SPC and MPEG-DSPE (PTX-NBD@PC-PEG NSs), PTX-NBD@PC-PEG NCs remarkably reduced burst release and improved cellular uptake efficiency and in vitro cancer cell killing ability. In a word, the work highlights the potential of theranostic prodrug nanocrystals based on the drug-fluorophore conjugates for cell imaging and chemotherapy.


Subject(s)
Nanoparticles/chemistry , Paclitaxel/chemistry , Phosphatidylcholines/chemistry , Phosphatidylethanolamines/chemistry , Polyethylene Glycols/chemistry , Prodrugs/chemistry , Cell Survival/drug effects , Fluorescence , HeLa Cells , Humans , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Nitro Compounds/chemistry , Oxadiazoles/chemistry , Paclitaxel/pharmacology , Prodrugs/pharmacology , Glycine max/chemistry , Spectroscopy, Fourier Transform Infrared , Theranostic Nanomedicine/methods , X-Ray Diffraction
20.
ACS Appl Mater Interfaces ; 9(40): 34650-34665, 2017 Oct 11.
Article in English | MEDLINE | ID: mdl-28920426

ABSTRACT

"All-in-one" carrier-free-based nano-multi-drug self-delivery system could combine triple advantages of small molecules, nanoscale characteristics, and synergistic combination therapy together. Researches have showed that dual-acting small-molecular methotrexate (MTX) could target and kill the folate-receptor-overexpressing cancer cells. Inspired by this mechanism, a novel collaborative early-phase tumor-selective targeting and late-phase synergistic anticancer approach was developed for the self-assembly of chemotherapeutic drug-drug conjugate, which showed various advantages of more simplicity, efficiency, and flexibility over the conventional approach based only on single or combination cancer chemotherapy. MTX and 10-hydroxyl camptothecin (CPT) were chosen to conjugate through ester linkage. Because of the amphiphilicity and ionicity, MTX-CPT conjugates as molecular building blocks could self-assemble into MTX-CPT nanoparticles (MTX-CPT NPs) in aqueous solution, thus notably improving the aqueous solubility of CPT and the membrane permeability of MTX. The MTX-CPT NPs with a precise drug-to-drug ratio showed pH-/esterase-responsive drug release, sequential function "Targeting-Anticancer" switch, and real-time monitoring fluorescence "Off-On" switch. By doping with a lipophilic near-infrared (NIR) cyanine dye (e.g., 1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide, DiR), the prepared DiR-loaded MTX-CPT NPs acted as an effective probe for in vivo NIR fluorescence (NIRF) and photoacoustic (PA) dual-modal imaging. Both in vitro and in vivo studies demonstrated that MTX-CPT NPs could specifically codeliver multidrug to different sites of action with distinct anticancer mechanisms to kill folate-receptor-overexpressing tumor cells in a synergistic way. This novel, simple, and highly convergent self-targeting nanomulti-drug codelivery system exhibited great potential in cancer therapy.


Subject(s)
Nanoparticles , Camptothecin , Cell Line, Tumor , Drug Delivery Systems , Humans , Methotrexate , Prodrugs
SELECTION OF CITATIONS
SEARCH DETAIL
...