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










Publication year range
1.
ACS Nano ; 18(26): 16674-16683, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38907991

ABSTRACT

Targeted nanoparticles have been extensively explored for their ability to deliver their payload to a selective cell population while reducing off-target side effects. The design of actively targeted nanoparticles requires the grafting of a ligand that specifically binds to a highly expressed receptor on the surface of the targeted cell population. Optimizing the interactions between the targeting ligand and the receptor can maximize the cellular uptake of the nanoparticles and subsequently improve their activity. Here, we evaluated how the density and presentation of the targeting ligands dictate the cellular uptake of nanoparticles. To do so, we used a DNA-scaffolded PLGA nanoparticle system to achieve efficient and tunable ligand conjugation. A prostate-specific membrane antigen (PSMA) expressing a prostate cancer cell line was used as a model. The density and presentation of PSMA targeting ligand ACUPA were precisely tuned on the DNA-scaffolded nanoparticle surface, and their impact on cellular uptake was evaluated. It was found that matching the ligand density with the cell receptor density achieved the maximum cellular uptake and specificity. Furthermore, DNA hybridization-mediated targeting chain rigidity of the DNA-scaffolded nanoparticle offered ∼3 times higher cellular uptake compared to the ACUPA-terminated PLGA nanoparticle. Our findings also indicated a ∼ 3.7-fold reduction in the cellular uptake for the DNA hybridization of the non-targeting chain. We showed that nanoparticle uptake is energy-dependent and follows a clathrin-mediated pathway. Finally, we validated the preferential tumor targeting of the nanoparticles in a bilateral tumor xenograft model. Our results provide a rational guideline for designing actively targeted nanoparticles and highlight the application of DNA-scaffolded nanoparticles as an efficient active targeting platform.


Subject(s)
DNA , Glutamate Carboxypeptidase II , Nanoparticles , Prostatic Neoplasms , Nanoparticles/chemistry , Humans , DNA/chemistry , DNA/metabolism , Ligands , Male , Glutamate Carboxypeptidase II/metabolism , Glutamate Carboxypeptidase II/chemistry , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Animals , Cell Line, Tumor , Mice , Antigens, Surface/metabolism , Antigens, Surface/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry
2.
ACS Nano ; 18(11): 8143-8156, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38436248

ABSTRACT

The complexity and heterogeneity of individual tumors have hindered the efficacy of existing therapeutic cancer vaccines, sparking intensive interest in the development of more effective in situ vaccines. Herein, we introduce a cancer nanovaccine for reactive oxygen species-augmented metalloimmunotherapy in which FeAl-layered double hydroxide (LDH) is used as a delivery vehicle with dihydroartemisinin (DHA) as cargo. The LDH framework is acid-labile and can be degraded in the tumor microenvironment, releasing iron ions, aluminum ions, and DHA. The iron ions contribute to aggravated intratumoral oxidative stress injury by the synergistic Fenton reaction and DHA activation, causing apoptosis, ferroptosis, and immunogenic cell death in cancer cells. The subsequently released tumor-associated antigens with the aluminum adjuvant form a cancer nanovaccine to generate robust and long-term immune responses against cancer recurrence and metastasis. Moreover, Fe ion-enabled T1-weighted magnetic resonance imaging can facilitate real-time tumor therapy monitoring. This cancer-nanovaccine-mediated metalloimmunotherapy strategy has the potential for revolutionizing the precision immunotherapy landscape.


Subject(s)
Artemisinins , Nanoparticles , Neoplasms , Humans , Reactive Oxygen Species/metabolism , Nanovaccines , Aluminum , Neoplasms/drug therapy , Iron , Hydroxides , Immunotherapy/methods , Tumor Microenvironment
3.
Adv Mater ; : e2206401, 2022 Oct 10.
Article in English | MEDLINE | ID: mdl-36210733

ABSTRACT

Multienzyme-mimicking redox nanozymes, curated by defect engineering, in synergy with immunotherapy offer promising prospects for safe and efficient cancer therapy. However, the spatiotemporally precise immune response often gets challenged by off-target adverse effects and insufficient therapeutic response. Herein, a tumor cell membrane coated redox nanozyme (CMO-R@4T1) is reported for combinational second near-infrared window (NIR-II) photothermal immunotherapy. CMO-R@4T1 consists of a Cu-doped MoOx (CMO) nanozyme as the core, which is cloaked with tumor-cell-derived fused membranes with immunostimulants immobilized in the membrane shell. In addition to the enhanced tumor accumulation, the nanozyme can cause oxidative damage to tumor cells by the production of reactive oxygen species and attenuation of the antioxidant mechanism. CMO-R@4T1 also mediates a photothermal effect under NIR-II photoirradiation to trigger tumor eradication and immunogenic cell death, where the liberated agonist elicits the immune activation. Such a controlled therapeutic paradigm potentiates systemic primary tumor ablation, inhibits cancer metastasis to distant tumor, and procures long-term immunological memory. Thereby, this study takes advantage of defect engineering to illustrate a generic strategy to prepare cell-membrane-camouflaged nanozymes for targeted photo-immunotherapy of cancer.

4.
Adv Mater ; 34(21): e2201706, 2022 May.
Article in English | MEDLINE | ID: mdl-35357041

ABSTRACT

Although photothermal immunotherapy (PTI) is a compelling strategy for tumor therapy, the development of promising photothermal agents to overcome the insufficient immunogenicity of tumor cells and the poor immune response encountered in PTI is still challenging. Herein, commercial small-molecule-based organic metal adjuvants (OMAs) are presented, with second near-infrared photoacoustic and photothermal properties as well as the ability to perturb redox homeostasis to potentiate immunogenicity and immune responsiveness. OMAs, assembled from charge-transfer complexes and characterized by a broad substrate scope, high accessibility, and flexibly tuned optical properties, demonstrate strong phototherapeutic and adjuvant abilities via the depletion of glutathione and cysteine, and subsequently elicit systemic immunity by evoking immunogenic cell death, promoting dendritic cell maturation, and increasing T cell infiltration. Furthermore, programmed cell death protein 1 antibody can be employed to synergize with OMAs to suppress tumor immune evasion and ultimately improve the treatment outcomes. This study unlocks new paradigms to provide a versatile OMA-based scaffold for future practical applications.


Subject(s)
Nanoparticles , Phototherapy , Adjuvants, Immunologic , Cell Line, Tumor , Glutathione , Immunotherapy , Nanoparticles/chemistry
5.
Exploration (Beijing) ; 2(2): 20210238, 2022 Apr.
Article in English | MEDLINE | ID: mdl-37323881

ABSTRACT

Chemodynamic therapy (CDT) has emerged to be a frontrunner amongst reactive oxygen species-based cancer treatment modalities. CDT utilizes endogenous H2O2 in tumor microenvironment (TME) to produce cytotoxic hydroxyl radicals (•OH) via Fenton or Fenton-like reactions. While possessing advantages such as tumor specificity, no need of external stimuli, and low side effects, practical applications of CDT are still impeded owing to the heterogeneity, complexity, and reductive environment of TME. Over the past couple of years, strategies to enhance CDT for efficient tumor regression are in rapid development in synergy with the growth of nanomedicine. In this review, we initially outline the fundamental understanding of Fenton and Fenton-like reactions and their relationship with CDT. Subsequently, the development in the design of nanosystems for CDT is highlighted in a general manner. Furthermore, recent advancement of the strategies to augment Fenton reactions in TME for enhanced CDT is discussed in detail. Finally, perspectives toward the future development of CDT for better therapeutic outcome are presented. This review is expected to draw attention for collaborative research on CDT in the best interest of its future clinical applications.

6.
Biomaterials ; 279: 121188, 2021 12.
Article in English | MEDLINE | ID: mdl-34678649

ABSTRACT

There is an impending need for the development of carrier-free nanosystems for single laser triggered activation of phototherapy, as such approach can overcome the drawbacks associated with irradiation by two distinct laser sources for avoiding prolonged treatment time and complex treatment protocols. Herein, we developed a self-assembled nanosystem (SCP-CS) consisting of a new semiconducting polymer (SCP) and encapsulated ultrasmall CuS (CS) nanoparticles. The SCP component displays remarkable near infrared (NIR) induced photothermal ability, enhanced reactive oxygen species (ROS) generation, and incredible photoacoustic (PA) signals upon activation by 808 nm laser for phototherapy mediated cancer ablation. The CuS component improves the PA imaging ability of SCP-CS, and also enhances photo-induced chemodynamic efficacy. Attributed to promoted single laser-triggered hyperthermia and enhanced ROS generation, the SCP-CS nanosystem shows effective intracellular uptake and intratumoral accumulation, enhanced tumor suppression with reduced treatment time, and devoid of any noticeable toxicity.


Subject(s)
Hyperthermia, Induced , Nanoparticles , Neoplasms , Humans , Neoplasms/therapy , Phototherapy , Polymers
7.
Angew Chem Int Ed Engl ; 60(50): 26254-26259, 2021 12 06.
Article in English | MEDLINE | ID: mdl-34591365

ABSTRACT

Clinical translation of artesunate (ATS) as a potent antitumor drug has been obstructed by its rapid degradation and low bioavailability. Herein, we report the development of an ATS nanomedicine through the self-assembly with Mn[Co(CN)6 ]2/3 □1/3 metal-organic frameworks (MOFs) that have hidden missing linkers. The defects in MOFs originating from the missing linkers play a key role in increasing the biological stability and tumor accumulation of ATS. Chlorin e6 (Ce6) and ATS can be co-loaded into MOFs for a synergistic antitumor efficacy. In the presence of intracellular HCO3- , Mn2+ acts as an efficient catalyst to promote the bicarbonate-activated H2 O2 system which oxidizes ATS to generate reactive oxygen species and induce oxidative death to cancer cells. The released [CoIII (CN)6 ] linker undergoes a redox reaction with intracellular glutathione to prevent the scavenging ability of reactive oxygen species, contributing to synergistic chemodynamic therapy of ATS and photodynamic therapy of Ce6. Thus, defect-engineered MOFs with hidden missing linkers hold great promise in advancing the practical use of ATS as an antitumor medicine.


Subject(s)
Antineoplastic Agents/pharmacology , Artesunate/pharmacology , Breast Neoplasms/drug therapy , Metal-Organic Frameworks/chemistry , Photosensitizing Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Artesunate/chemistry , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Carriers/chemistry , Drug Screening Assays, Antitumor , Female , Humans , Mammary Neoplasms, Experimental/diagnostic imaging , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Photosensitizing Agents/chemistry , Reactive Oxygen Species/metabolism
8.
ACS Nano ; 15(4): 7774-7782, 2021 04 27.
Article in English | MEDLINE | ID: mdl-33844517

ABSTRACT

The therapeutic effect of chemodynamic therapy (CDT) is significantly restricted by the stern reaction conditions and slow reaction rate of the Fenton reaction (pH 3-4). Herein, we report an ultrasmall trimetallic (Pd, Cu, and Fe) alloy nanozyme (PCF-a NEs) possessing dynamic active-site synergism, thus exhibiting a cascade glutathione peroxidase and peroxidase (POD) mimicking activities in circumneutral pH. PCF-a NEs exhibit photothermally augmented POD property and high photothermal conversion efficiency (62%) for synergistic tumor cell apoptosis. In addition, ultrasound can also enhance the mass transfer at active catalytic sites of PCF-a NEs, in turn accelerating Fenton-like reaction for tumor-specific CDT. This work provides a strategy for engineering alloy nanozymes in a bioinspired way for the amplification of intratumor reactive oxygen species in response to external stimuli, demonstrating enhanced efficiency for the inhibition of tumor growth in vitro and in vivo.


Subject(s)
Alloys , Neoplasms , Catalysis , Humans , Infrared Rays , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Reactive Oxygen Species
9.
JACS Au ; 1(12): 2328-2338, 2021 Dec 27.
Article in English | MEDLINE | ID: mdl-34977901

ABSTRACT

The efficacy of reactive oxygen species (ROS)-based therapy is substantially constrained by the limited ROS generation, stern activation conditions, and lack of a straightforward reaction paradigm. Carbon dots (CDs) have been highly sought after for therapeutic applications for their biocompatibility and intrinsic fluorescence imaging capabilities, making them suitable for ROS generation. Herein, we synthesized a CD-based ultrasmall hybrid nanostructure possessing active sites of Mo, Cu, and IR-780 dye. After cooperative self-assembly with 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-poly(ethylene glycol), the obtained assembly (CMIR-CDa) exhibits near-infrared fluorescence imaging and photoacoustic tomography. Interestingly, CMIR-CDa can generate singlet oxygen (1O2), hydroxyl radical (·OH), and superoxide radical anion (O2 • -) upon ultrasound stimulus owing to its sonosensitizing and enzyme-mimicking properties, showing an enhanced efficacy for tumor ablation in vivo. The collective in vitro and in vivo results indicate that CMIR-CDa has a high potency as an ROS nanogenerator under US irradiation, even at a low concentration. The present study offers an approach for engineering hybrid CDs in a bioinspired way for intratumoral ROS augmentation in response to deep tissue penetrable external stimuli.

10.
Angew Chem Int Ed Engl ; 60(6): 3001-3007, 2021 02 08.
Article in English | MEDLINE | ID: mdl-33091204

ABSTRACT

Nanomaterials with enzyme-mimicking activity (nanozymes) show potential for therapeutic interventions. However, it remains a formidable challenge to selectively kill tumor cells through enzymatic reactions, while leaving normal cells unharmed. Herein, we present a new strategy based on a single-site cascade enzymatic reaction for tumor-specific therapy that avoids off-target toxicity to normal tissues. A copper hexacyanoferrate (Cu-HCF) nanozyme with active single-site copper exhibited cascade enzymatic activity within the tumor microenvironment: Tumor-specific glutathione oxidase activity by the Cu-HCF single-site nanozymes (SSNEs) led to the depletion of intracellular glutathione and the conversion of single-site CuII species into CuI for subsequent amplified peroxidase activity through a Fenton-type Harber-Weiss reaction. In this way, abundant highly toxic hydroxyl radicals were generated for tumor cell apoptosis. The results show that SSNEs could amplify the tumor-killing efficacy of reactive oxygen species and suppress tumor growth in vivo.


Subject(s)
Biomimetic Materials/chemistry , Copper/chemistry , Ferrocyanides/chemistry , Nanostructures/chemistry , Animals , Biomimetic Materials/metabolism , Catalysis , Cell Line , Cell Survival/drug effects , Glutathione/chemistry , Glutathione/metabolism , Humans , Hydrogen Peroxide/chemistry , Hydroxyl Radical/chemistry , Hydroxyl Radical/metabolism , Iron/chemistry , Mice , Microscopy, Confocal , Nanostructures/therapeutic use , Nanostructures/toxicity , Neoplasms/drug therapy , Neoplasms/pathology , Oxidoreductases/chemistry , Oxidoreductases/metabolism , Reactive Oxygen Species/chemistry , Reactive Oxygen Species/metabolism
11.
ACS Nano ; 14(10): 13500-13511, 2020 10 27.
Article in English | MEDLINE | ID: mdl-32910637

ABSTRACT

Intracellular antioxidants such as glutathione (GSH) play a critical role in protecting malignant tumor cells from apoptosis induced by reactive oxygen species (ROS) and in mechanisms of multidrug and radiation resistance. Herein, we rationally design two multicomponent self-assembled photodynamic therapy (PDT) nanoagents, that is, Glup-MFi-c and Glud-MFo-c, which consist of respective GSH-passivation and GSH-depletion linkers in metal-organic frameworks encapsulated with photosensitizers for a deeply comprehensive understanding of GSH-based tumor PDT. Multicomponent coordination, π-π stacking, and electrostatic interactions among metal ions, photosensitizers, and bridging linkers under the protection of a biocompatible polymer generate homogeneous nanoparticles with satisfied size, good colloid stability, and ultrahigh loading capacity. Compared to the GSH-passivated Glup-MFi-c, the GSH-depleted Glud-MFo-c shows pH-responsive release of photosensitizer and [FeIII(CN)6] linker in tumor cells to efficiently deplete intracellular GSH, thus amplifying the cell-killing efficiency of ROS and suppressing the tumor growth in vivo. This study demonstrates that Glud-MFo-c acts as a ROS amplifier, providing a useful strategy to deeply understand the role of GSH in combating cancer.


Subject(s)
Metal-Organic Frameworks , Photochemotherapy , Ferric Compounds , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species
12.
Acc Chem Res ; 53(7): 1389-1400, 2020 07 21.
Article in English | MEDLINE | ID: mdl-32597637

ABSTRACT

Nanozymes, which integrate the advantages of both nanomaterials and natural enzymes, have accumulated enormous research interest over the past decades because of the opportunity they provide to appreciate and further cultivate artificial enzymes with comparable properties. By mimicking the coordination environments of the catalytic sites in natural enzymes, nanozymes with confined nanostructures can serve as substitutes in many catalytic processes with comparable activity and robust stability even in harsh conditions. Since the pioneering report about peroxidase-mimicking ferromagnetic nanoparticles in 2007, nanozymes have been developed as specialized for nanomaterials with intrinsic enzyme-mimicking property. With the rapid development in nanoscience and nanotechnology, nanomaterials with superior advantages, such as large-scale production, desired activity, and robust stability, can bridge the natural enzymes with nanozymes.Metal-organic frameworks (MOFs) and their derivatives hold great promise to serve as direct surrogates of conventional enzymes for enzymatic reactions. According to their chemical nature, MOF-based nanozymes can be divided into three main categories: pristine MOFs, enzyme-encapsulated MOF composites, and MOF-based derivatives. Due to the versatility of metallic nodes and bridging linkers together with the feasibility of postsynthetic engineering and modification, MOFs and their derivatives are envisioned as one of the most appropriate surrogates for this purpose. Using MOFs as precursors or sacrificial templates, multiple MOF-based derivatives including carbon-based nanomaterials (e.g., heteroatom-doped carbon or carbon with M-N-C moiety), metal oxide/carbon nanoparticles, and metal/carbon nanomaterials can be rationally synthesized through one-step direct carbonization/oxidation or indirect post-synthesis treatments of MOFs (e.g., bridging linker-exchange and metallic node-doping). Compared with existing nanozymes, MOF-based derivatives open up a new avenue for constructing mesoporous nanozymes. In this way, the intrinsic mesoporous properties of MOFs can still be maintained, while the stability and activity can be greatly improved. In this Account, we highlight some important research advances in MOF-based derivatives (including M-N-C moieties (M = single metal atom), metal oxide/carbon, metal/carbon, and MOF derivatives obtained through postsynthetic linker exchange and metal doping strategies) with enzyme-mimicking activity. We also demonstrate that, through integrating physicochemical properties of mesoporous nanomaterials and enzymatic activities of natural enzymes, MOF-derived nanozymes can provide multifunctional platforms in biomedical fields such as antibacterial agents, biosensors, imaging, cancer therapy, and environmental protection. Finally, we propose future design principles and possible research approaches for deeper understanding of mechanisms, thus pointing out future research directions to offer more opportunities for the conventional enzyme-engineering industry.


Subject(s)
Enzymes/chemistry , Metal-Organic Frameworks/chemistry , Nanostructures , Medicine
13.
Chem Asian J ; 15(16): 2513-2519, 2020 Aug 17.
Article in English | MEDLINE | ID: mdl-32583605

ABSTRACT

Development of heterogeneous catalysts has attracted increasing attention, owing to their remarkable catalytic performance and recyclability. Herein, we report well-developed heterogeneous catalysts with a three-dimensional ordered hierarchical structure, constructed from nickel or cobalt nanoparticles embedded in porous carbon. The obtained catalysts were fully characterized by several techniques. On account of the uniform distribution of metal nanoparticles in the porous carbon matrix and large diffusion channels that allow for effective mass transport, the catalysts exhibited superior catalytic performance for styrene epoxidation reaction. In particular, the catalysts showed good catalytic activity, high selectivity and excellent recyclability toward the styrene epoxidation. Thus, this facile approach developed allows for fabricating advanced heterogeneous catalysts with high catalytic activities for useful practical applications.

14.
Angew Chem Int Ed Engl ; 59(28): 11287-11292, 2020 Jul 06.
Article in English | MEDLINE | ID: mdl-32250502

ABSTRACT

Photocatalysts derived from semiconductor heterojunctions that harvest solar energy and catalyze reactions still suffer from low solar-to-hydrogen conversion efficiency. Now, MXene (Ti3 C2 TX ) nanosheets (MNs) are used to support the in situ growth of ultrathin ZnIn2 S4 nanosheets (UZNs), producing sandwich-like hierarchical heterostructures (UZNs-MNs-UZNs) for efficient photocatalytic H2 evolution. Opportune lateral epitaxy of UZNs on the surface of MNs improves specific surface area, pore diameter, and hydrophilicity of the resulting materials, all of which could be beneficial to the photocatalytic activity. Owing to the Schottky junction and ultrathin 2D structures of UZNs and MNs, the heterostructures could effectively suppress photoexcited electron-hole recombination and boost photoexcited charge transfer and separation. The heterostructure photocatalyst exhibits improved photocatalytic H2 evolution performance (6.6 times higher than pristine ZnIn2 S4 ) and excellent stability.

15.
ACS Appl Mater Interfaces ; 12(16): 18342-18351, 2020 Apr 22.
Article in English | MEDLINE | ID: mdl-32223204

ABSTRACT

Therapeutic efficacy of synergistic photodynamic therapy (PDT) and photothermal therapy (PTT) is limited by complex conjugation chemistry, absorption wavelength mismatch, and inadequate biodegradability of the PDT-PTT agents. Herein, we designed biocompatible copper sulfide nanodot anchored folic acid-modified black phosphorus nanosheets (BP-CuS-FA) to overcome these limitations, consequently enhancing the therapeutic efficiency of PDT-PTT. In vitro and in vivo assays reveal good biocompatibility and commendable tumor inhibition efficacy of the BP-CuS-FA nanoconjugate because of the synergistic PTT-PDT mediated by near-infrared laser irradiation. Importantly, folic acid unit could target folate receptor overexpressed cancer cells, leading to enhanced cellular uptake of BP-CuS-FA. BP-CuS-FA also exhibits significant contrast effect for photoacoustic imaging, permitting its in vivo tracking. The photodegradable character of BP-CuS-FA is associated with better renal clearance after the antitumor therapy in vivo. The present research may facilitate further development on straightforward approaches for targeted and imaging-guided synergistic PDT-PTT of cancer.


Subject(s)
Antineoplastic Agents , Nanoconjugates/chemistry , Phosphorus , Photoacoustic Techniques/methods , Phototherapy/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cell Survival/drug effects , Copper Sulfate/chemistry , Female , Folic Acid/chemistry , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/therapy , Phosphorus/chemistry , Phosphorus/pharmacokinetics , Phosphorus/pharmacology , Phosphorus/therapeutic use , Theranostic Nanomedicine/methods
17.
Adv Mater ; 31(25): e1901513, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31069885

ABSTRACT

Uncontrolled cancer cell proliferation, insufficient blood flow, and inadequate endogenous oxygen lead to hypoxia in tumor tissues. Herein, a unique type of hypoxia-responsive human serum albumin (HSA)-based nanosystem (HCHOA) is reported, prepared by cross-linking the hypoxia-sensitive azobenzene group between photosensitizer chlorin e6 (Ce6)-conjugated HSA (HC) and oxaliplatin prodrug-conjugated HSA (HO). The HCHOA nanosystem is stable under normal oxygen partial pressure with a size of 100-150 nm. When exposed to the hypoxic tumor microenvironment, the nanosystem can quickly dissociate into ultrasmall HC and HO therapeutic nanoparticles with a diameter smaller than 10 nm, significantly enabling their enhanced intratumoral penetration. After the dissociation, the quenched fluorescence of Ce6 in the produced HC nanoparticles can be recovered for bioimaging. At the same time, the production of singlet oxygen is increased because of the enhancement in the photoactivity of the photosensitizer. On account of these improvements, combined photodynamic therapy and chemotherapy is realized to display superior antitumor efficacy in vivo. Based on this simple strategy, it is possible to achieve the dissociation of hypoxic-responsive nanosystem to enhance the tumor penetration and therapeutic effect.


Subject(s)
Drug Carriers/chemistry , Drug Carriers/metabolism , Nanoparticles/chemistry , Serum Albumin, Human/chemistry , Serum Albumin, Human/metabolism , Tumor Hypoxia , Animals , Cell Line, Tumor , Chlorophyllides , Humans , Mice , Oxaliplatin/chemistry , Oxaliplatin/metabolism , Oxaliplatin/pharmacology , Porphyrins/chemistry , Porphyrins/pharmacology , Prodrugs/metabolism , Tumor Hypoxia/drug effects
18.
Sci Adv ; 4(5): eaas9732, 2018 05.
Article in English | MEDLINE | ID: mdl-29736419

ABSTRACT

Ultralong room temperature phosphorescence (URTP) emitted from pure amorphous organic molecules is very rare. Although a few crystalline organic molecules could realize URTP with long lifetimes (>100 ms), practical applications of these crystalline organic phosphors are still challenging because the formation and maintenance of high-quality crystals are very difficult and complicated. Herein, we present a rational design for minimizing the vibrational dissipation of pure amorphous organic molecules to achieve URTP. By using this strategy, a series of URTP films with long lifetimes and high phosphorescent quantum yields (up to 0.75 s and 11.23%, respectively) were obtained from amorphous organic phosphors without visible fluorescence and phosphorescence under ambient conditions. On the basis of the unique features of URTP films, a new green screen printing technology without using any ink was developed toward confidential information encryption and decryption. This work presents a breakthrough strategy in applying amorphous organic materials for URTP.

SELECTION OF CITATIONS
SEARCH DETAIL
...