Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 86
Filter
1.
J Craniofac Surg ; 33(4): 1130-1135, 2022 Jun 01.
Article in English | MEDLINE | ID: mdl-34560753

ABSTRACT

ABSTRACT: The expanded pedicled deltopectoral flap (EPDF) has been widely used to repair large facial scars. Although doctors and patients are usually satisfied with the outcomes, the actual functional recovery and cosmetic effects of EPDF are still unknown. It is, therefore, necessary to objectively investigate the effect of transferred EPDF by using a variety of methods. From January 2008 to December 2018, 52 patients who underwent EPDF surgery at Xijing Hospital were enrolled. Sense of touch, static 2-point discrimination, elasticity, and color were measured. Thermesthesia and algesthesia were also tested. Postoperative scars were evaluated using the patient and observer scar assessment scale. Satisfaction of patients, doctors, and laypersons was investigated. The algaesthesis, thalposis, and rhigosis scores were 4.7 ±â€Š0.7, 3.7 ±â€Š0.9, and 4.5 ±â€Š0.8, respectively. The tactile score was 0.3 ±â€Š0.2 mN, and 2-point discrimination was 10.1 ±â€Š4.8 mm. L ∗ , a ∗ hemoglobin, and melanin content of the flaps were significantly different when compared with normal skin ( P   <  0.05). The satisfaction of doctors, patients, and laypersons was 88.5%, 71.2%, and 67.3%, respectively. The higher satisfaction of patients was mainly associated with the smaller color difference between the flap and the surrounding skin, and lower patient and observer scar assessment scale score. These results confirm that excellent functional recovery and reliable cosmetic effects are observed when facial scars are repaired with EPDF. The methods used in this study can be applied to the evaluation of functional recovery and cosmetic outcomes of transferred flaps, which may provide a more comprehensive understanding of flap assessment.


Subject(s)
Cicatrix , Face , Plastic Surgery Procedures , Surgical Flaps , Cicatrix/surgery , Face/surgery , Humans , Plastic Surgery Procedures/methods , Skin , Skin Transplantation/methods , Surgical Flaps/surgery , Treatment Outcome
2.
J Am Chem Soc ; 142(14): 6822-6832, 2020 04 08.
Article in English | MEDLINE | ID: mdl-32196319

ABSTRACT

Biocatalytic reactions in living cells involve complex transformations in the spatially confined microenvironments. Inspired by biological transformation processes, we demonstrate effective biocatalytic cascade driven photodynamic therapy in tumor-bearing mice by the integration of an artificial enzyme (ultrasmall Au nanoparticles) with upconversion nanoparticles (NaYF4@NaYb0.92F4:Er0.08@NaYF4)zirconium/iron porphyrin metal-organic framework core-shell nanoparticles (UMOF NPs) which act as biocatalysts and nanoreactors. The construction of core-shell UMOF NPs are realized by using a unique "solvent-assisted self-assembly" method. The integration of ultrasmall AuNPs on the UMOFs matrix leads to glucose depletion, providing Au-mediated cancer therapy via glucose oxidase like catalytic activity. Meanwhile, the UMOF matrix acts as a near-infrared (NIR) light photon-activated singlet oxygen generator through a continuous supply of oxygen via hydrogen peroxide decomposition upon irradiation. Such kinds of biocatalysts offer exciting opportunities for biomedical, catalytical ,and energy applications.


Subject(s)
Metal Nanoparticles/chemistry , Metal-Organic Frameworks/metabolism , Photochemotherapy/methods , Humans
3.
Adv Funct Mater ; 30(4)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-33041745

ABSTRACT

The combination of reactive oxygen species (ROS)-involved photodynamic therapy (PDT) and chemodynamic therapy (CDT) holds great promise for enhancing ROS-mediated cancer treatment. Herein, we reported an in situ polymerized hollow mesoporous organosilica nanoparticle (HMON) biocatalysis nanoreactor to integrate the synergistic effect of PDT/CDT for enhancing ROS-mediated pancreatic ductal adenocarcinoma treatment. HPPH photosensitizer was hybridized within the framework of HMON via an "in situ framework growth" approach. Then, the hollow cavity of HMONs was exploited as a nanoreactor for "in situ polymerization" to synthesize the polymer containing thiol groups, thereby enabling the immobilization of ultrasmall gold nanoparticles, which behave like glucose oxidase-like nanozyme, converting glucose into H2O2 to provide self-supplied H2O2 for CDT. Meanwhile, Cu2+-tannic acid complexes were further deposited on the surface of HMONs (HMON-Au@Cu-TA) to initiate Fenton-like reaction to covert the self-supplied H2O2 into •OH, a highly toxic ROS. Finally, collagenase (Col), which can degrade the collagen I fiber in the extracellular matrix (ECM), was loaded into HMON-Au@Cu-TA to enhance the penetration of HMONs and O2 infiltration for enhanced PDT. This study provides a good paradigm for enhancing ROS-mediated anti-tumor efficacy. Meanwhile, this research offers a new method to broaden the application of silica based nanotheranostics.

4.
Small ; 16(42): e2004016, 2020 10.
Article in English | MEDLINE | ID: mdl-32985099

ABSTRACT

Chemical transformation in cellular environment is critical for regulating biological processes and metabolic pathways. Harnessing biocatalytic cascades to produce chemicals of interest has become a research focus to benefit industrial and pharmaceutic areas. Nanoreactors, which can act as artificial cell-like devices to organize cascade reactions, have been recently proposed for potential therapeutic applications for life-threatening illnesses. Among various types of nanomaterials, there is a growing interest in 2D metal-organic frameworks (MOFs). By virtue of the ultralarge specific surface area, high porosity, and structural diversity, 2D MOF nanosheets hold great promise for a broad spectrum of biomedical use. Herein, a unique planar MOF-based hybrid architecture (GMOF-LA) is introduced by incorporating ultrasmall gold nanoparticles (Au NPs) as nanozyme and l-Arginine (l-Arg) as nitric oxide (NO) donor. The prepared Au NPs enable oxidation of glucose into hydrogen peroxide, which drives biocatalytic cascades to covert l-Arg into NO. Interestingly, the well-designed nanosheets not only possess excellent catalytical activity for NO generation, resulting in gas therapeutic effect, but also serve as a desired photosensitizer for photodynamic therapy. This study establishes a good example of exploring bioinspired nanoreactors for cooperative anticancer effect, which may pave the path for future "bench-to-bedside" design of nanomedicine.


Subject(s)
Metal Nanoparticles , Metal-Organic Frameworks , Neoplasms , Catalysis , Gold , Humans , Neoplasms/drug therapy
5.
J Am Chem Soc ; 141(37): 14687-14698, 2019 09 18.
Article in English | MEDLINE | ID: mdl-31466436

ABSTRACT

Phototheranostics refers to advanced photonics-mediated theranostic methods for cancer and includes imaging-guided photothermal/chemotherapy, photothermal/photodynamic therapy, and photodynamic/chemotherapy, which are expected to provide a paradigm of modern precision medicine. In this regard, various phototheranostic drug delivery systems with excellent photonic performance, controlled drug delivery/release, and precise photoimaging guidance have been developed. In this study, we reported a special "in situ framework growth" method to synthesize novel phototheranostic hollow mesoporous nanoparticles by ingenious hybridization of perylene diimide (PDI) within the framework of small-sized hollow mesoporous organosilica (HMO). The marriage of PDI and HMO endowed the phototheranostic silica nanoparticles (HMPDINs) with largely amplified fluorescence and photoacoustic signals, which can be used for enhanced fluorescence and photoacoustic imaging. The organosilica shell can be chemically chelated with isotope 64Cu for positron emission tomography imaging. Moreover, in situ polymer growth was introduced in the hollow structure of the HMPDINs to produce thermosensitive polymer (TP) in the cavity of HMPDINs to increase the loading capacity and prevent unexpected leakage of the hydrophobic drug SN38. Furthermore, the framework-hybridized PDI generated heat under near-infrared laser irradiation to trigger the deformation of TP for controlled drug release in the tumor region. The fabricated hybrid nanomedicine with organic-inorganic characteristic not only increases the cancer theranostic efficacy but also offers an attractive solution for designing powerful theranostic platforms.


Subject(s)
Imides/chemistry , Nanoparticles/chemistry , Organosilicon Compounds/chemistry , Perylene/chemistry , Precision Medicine , Theranostic Nanomedicine , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude , Polymerization , Porosity , Xenograft Model Antitumor Assays
6.
Small ; 15(16): e1900691, 2019 04.
Article in English | MEDLINE | ID: mdl-30913380

ABSTRACT

Activatable imaging probes are promising to achieve increased signal-to-noise ratio for accurate tumor diagnosis and treatment monitoring. Magnetic resonance imaging (MRI) is a noninvasive imaging technique with excellent anatomic spatial resolution and unlimited tissue penetration depth. However, most of the activatable MRI contrast agents suffer from metal ion-associated potential long-term toxicity, which may limit their bioapplications and clinical translation. Herein, an activatable MRI agent with efficient MRI performance and high safety is developed for drug (doxorubicin) loading and tumor signal amplification. The agent is based on pH-responsive polymer and gadolinium metallofullerene (GMF). This GMF-based contrast agent shows high relaxivity and low risk of gadolinium ion release. At physiological pH, both GMF and drug molecules are encapsulated into the hydrophobic core of nanoparticles formed by the pH-responsive polymer and shielded from the aqueous environment, resulting in relatively low longitudinal relativity and slow drug release. However, in acidic tumor microenvironment, the hydrophobic-to-hydrophilic conversion of the pH-responsive polymer leads to amplified MR signal and rapid drug release simultaneously. These results suggest that the prepared activatable MRI contrast agent holds great promise for tumor detection and monitoring of drug release.


Subject(s)
Drug Carriers , Fullerenes/chemistry , Gadolinium/chemistry , Magnetic Resonance Imaging , Animals , Contrast Media/chemistry , Delayed-Action Preparations/therapeutic use , Doxorubicin/chemistry , Drug Delivery Systems , Drug Liberation , HeLa Cells , Humans , Hydrogen-Ion Concentration , Mice , Nanoparticles/chemistry , Neoplasms/drug therapy , Polymers/chemistry , Tumor Microenvironment
7.
Bioconjug Chem ; 30(6): 1821-1829, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31117347

ABSTRACT

Magnetic resonance imaging (MRI) diagnosis is better assisted by contrast agents that can augment the signal contrast in the imaging appearance. However, this technique is still limited by the inherently low sensitivity on the recorded signal changes in conventional T1 or T2 MRI in a qualitative manner. Here, we provide a new paradigm of MRI diagnosis using T1- T2 dual-modal MRI contrast agents for contrast-enhanced postimaging computations on T1 and T2 relaxation changes. An albumin-binding molecule (i.e., truncated Evans blue) chelated with paramagnetic manganese ion was developed as a novel T1- T2 dual-modal MRI contrast agent at high magnetic field (7 T). Furthermore, the postimaging computations on T1- T2 dual-modal MRI led to greatly enhanced signal-to-noise ratios (SNR) and contrast-to-noise ratios (CNR) in both subcutaneous and orthotopic brain tumor models compared with traditional MRI methods. The T1- T2 dual-modal MRI computations have great potential to eliminate suspicious artifacts and false-positive signals in mouse brain imaging. This study may open new avenues for contrast-enhanced MRI diagnosis and holds great promise for precision medicine.


Subject(s)
Albumins/metabolism , Brain Neoplasms/diagnostic imaging , Contrast Media , Magnetic Resonance Imaging/methods , Animals , Humans , Mice , Sensitivity and Specificity
8.
Bioconjug Chem ; 30(6): 1745-1753, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31181890

ABSTRACT

As highly expressed in insulinomas, the glucagon-like peptide-1 receptor (GLP-1R) is believed to be an attractive target for diagnosis, localization, and treatment with radiolabeled exendin 4. However, the high and persistent radioactivity accumulation of exendin 4 in the kidneys limits accurate diagnosis and safe, as well as effective, radiotherapy in insulinomas. In this study, we intend to reduce the renal accumulation of radiolabeled exendin 4 through degradation mediated by brush border membrane enzymes. A new exendin 4 ligand NOTA-MVK-Cys40-Leu14-Exendin 4 containing Met-Val-Lys (MVK) linker between the peptide and 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA) chelator was synthesized and labeled with 68Ga. The in vitro mouse serum stability and cell binding affinity of the tracer were evaluated. Initial in vitro cleavage of the linker was determined by incubation of a model compound Boc-MVK-Dde with brush border membrane vesicles (BBMVs) with and without the inhibitor of neutral endopeptidase (NEP). Further cleavage studies were performed with the full structure of NOTA-MVK-Cys40-Leu14-Exendin 4. Kidney and urine samples were collected in the in vivo metabolism study after intravenous injection of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4. The microPET images were acquired in INS-1 tumor model at different time points; the radioactivity uptake of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 in tumor and kidneys were determined and compared with the control radiotracer without MVK linker. 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 was stable in mouse serum. The MVK modification did not affect the affinity of NOTA-MVK-Cys40-Leu14-Exendin 4 toward GLP-1R. The in vitro cleavage study and in vivo metabolism study confirmed that the MVK sequence can be recognized by BBM enzymes and cleaved at the amide bond between Met and Val, thus releasing the small fragment containing Met. MicroPET images showed that the tumor uptake of 68Ga-NOTA-MVK-Cys40-Leu14-Exendin 4 was comparable to that of the control, while the kidney uptake was significantly reduced. As a result, more favorable tumor to kidney ratios were achieved. In this study, a novel exendin 4 analogue, NOTA-MVK-Cys40-Leu14-Exendin 4, was successfully synthesized and labeled with 68Ga. With the cleavable MVK sequence, this ligand could be cleaved by the enzymes on kidneys, and releasing the fragment of 68Ga-NOTA-Met-OH, which will rapidly excrete from urine. As the high and consistent renal radioactivity accumulation could be significantly reduced, NOTA-MVK-Cys40-Leu14-Exendin 4 shows great potential in the diagnosis and radiotherapy for insulinoma.


Subject(s)
Exenatide/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Insulinoma/diagnostic imaging , Animals , Exenatide/chemistry , Exenatide/therapeutic use , Female , Gallium Radioisotopes/chemistry , Gallium Radioisotopes/therapeutic use , Glucagon-Like Peptide-1 Receptor/analysis , HEK293 Cells , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/therapeutic use , Humans , Insulinoma/radiotherapy , Mice , Positron-Emission Tomography , Theranostic Nanomedicine
9.
Bioconjug Chem ; 30(6): 1711-1723, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31082207

ABSTRACT

The effectiveness of numerous molecular drugs is hampered by their poor pharmacokinetics. Different from previous approaches with limited effectiveness, most recently, emerging high-affinity albumin binding moieties (ABMs) for in vivo hitchhiking of endogenous albumin opens up an avenue to chaperone small molecules for long-acting therapeutics. Although several FDA-approved fatty acids have shown prolonged residence and therapeutic effect, an easily synthesized, water-soluble, and high-efficiency ABM with versatile drug loading ability is urgently needed to improve the therapeutic efficacy of short-lived constructs. We herein identified an ideal bivalent Evans blue derivative, denoted as N(tEB)2, as a smart ABM-delivery platform to chaperone short-lived molecules, through both computational modeling screening and efficient synthetic schemes. The optimal N(tEB)2 could reversibly link two molecules of albumin through its two binding heads with a preferable spacer, resulting in significantly extended circulation half-life of a preloaded cargo and water-soluble. Notably, this in situ dimerization of albumin was able to sandwich peptide therapeutics to protect them from proteolysis. As an application, we conjugated N(tEB)2 with exendin-4 for long-acting glucose control in a diabetic mouse model, and it was superior to both previously tested NtEB-exendin-4 (Abextide) and the newly FDA-approved semaglutide, which has been arguably the best commercial weekly formula so far. Hence, this novel albumin binder has excellent clinical potential for next-generation biomimetic drug delivery systems.


Subject(s)
Evans Blue/analogs & derivatives , Evans Blue/metabolism , Exenatide/analogs & derivatives , Exenatide/metabolism , Serum Albumin/metabolism , Animals , Binding Sites , Cell Line, Tumor , Evans Blue/chemical synthesis , Exenatide/blood , Exenatide/chemical synthesis , Humans , Hypoglycemic Agents/blood , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/metabolism , Mice , Models, Molecular , Pharmaceutical Preparations/blood , Pharmaceutical Preparations/chemical synthesis , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/metabolism , Protein Binding , Protein Multimerization , Proteolysis , Rats , Serum Albumin/chemistry
10.
Bioorg Med Chem ; 27(19): 115041, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31402203

ABSTRACT

The efficient radiosynthesis of biomolecules utilizing minute quantities of maleimide substrate is important for availability of novel peptide molecular imaging agents. We evaluated both 3-18F-fluoropropane-1-thiol and 2-(2-(2-(2-18F-fluoroethoxy)ethoxy)ethoxy)ethane-1-thiol (18F-fluoro-PEG4 thiol) as prosthetic groups for radiolabeling under physiological conditions. The precursor employed a benzoate for protection of the thiol and an arylsulfonate leaving group. The radiofluorination was fully automated on an Eckert & Ziegler synthesis system using standard Kryptofix222/K2CO3 conditions. In order to minimize the amount of biological molecule required for subsequent conjugation, the intermediates, S-(3-18F-fluoropropyl) benzothioate and 18F-fluoro-PEG4 benzothioate, were purified by HPLC. The intermediates were isolated from the HPLC in yields of 37-47% and 28-35%, respectively, and retrieved from eluate using solid phase extraction. Treatment of the benzothioates with sodium methoxide followed by acetic acid provided the free thiols. The desired maleimide substrate in acetonitrile or phosphate buffer was then added and incubated at room temperature for 15 min. The final radiolabeled bioconjugate was purified on a separate HPLC or NAP-5 column. Maleimides utilized for the coupling reaction included phenyl maleimide, an Evans Blue maleimide derivative, a dimeric RGDfK maleimide (E[c(RGDfK)]2), two aptamer maleimides, and PSMA maleimide derivative. Isolated radiochemical yields (non-decay corrected) of maleimide addition products based on starting 18F-fluoride ranged from 6 to 22% in a synthesis time of about 90 min. 18F-thiol prosthetic groups were further tested in vivo by conjugation to E[c(RGDfK)]2 maleimide in a U87MG xenograft model. PET studies demonstrated similar tumor accumulation of both prosthetic groups. 18F-fluoro-PEG4-S-E[c(RGDfK)]2 displayed a somewhat favorable pharmacokinetics compared to 18F-fluoropropyl-S-E[c(RGDfK)]2. Bone uptake was low for both indicating in vivo stability.


Subject(s)
Alkanesulfonates/chemistry , Indicators and Reagents/chemistry , Maleimides/pharmacology , Peptides/pharmacology , Radiopharmaceuticals/pharmacology , Sulfhydryl Compounds/chemistry , Animals , Cell Line, Tumor , Fluorine Radioisotopes/chemistry , Humans , Isotope Labeling/methods , Maleimides/chemical synthesis , Mice, Nude , Peptides/chemical synthesis , Radiopharmaceuticals/chemical synthesis
11.
Angew Chem Int Ed Engl ; 58(41): 14758-14763, 2019 10 07.
Article in English | MEDLINE | ID: mdl-31429173

ABSTRACT

Reactive oxygen species (ROS) can be used not only as a therapeutic agent for chemodynamic therapy (CDT), but also as a stimulus to activate release of antitumor drugs, achieving enhanced efficacy through the combination of CDT and chemotherapy. Here we report a pH/ROS dual-responsive nanomedicine consisting of ß-lapachone (Lap), a pH-responsive polymer, and a ROS-responsive polyprodrug. In the intracellular acidic environment, the nanomedicine can realize pH-triggered disassembly. The released Lap can efficiently generate hydrogen peroxide, which will be further converted into highly toxic hydroxyl radicals via the Fenton reaction. Subsequently, through ROS-induced cleavage of thioketal linker, doxorubicin is released from the polyprodrug. In vivo results indicate that the cascade of ROS generation and antitumor-drug release can effectively inhibit tumor growth. This design of nanomedicine with cascade reactions offers a promising strategy to enhance antitumor efficacy.


Subject(s)
Antibiotics, Antineoplastic/chemistry , Doxorubicin/chemistry , Naphthoquinones/chemistry , Prodrugs , A549 Cells , Animals , Drug Liberation , Humans , Mice , Nanoparticles , Neoplasms, Experimental/drug therapy , Reactive Oxygen Species
12.
Angew Chem Int Ed Engl ; 58(26): 8752-8756, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31046176

ABSTRACT

Tumor hypoxia, the "Achilles' heel" of current cancer therapies, is indispensable to drug resistance and poor therapeutic outcomes especially for radiotherapy. Here we propose an in situ catalytic oxygenation strategy in tumor using porphyrinic metal-organic framework (MOF)-gold nanoparticles (AuNPs) nanohybrid as a therapeutic platform to achieve O2 -evolving chemoradiotherapy. The AuNPs decorated on the surface of MOF effectively stabilize the nanocomposite and serve as radiosensitizers, whereas the MOF scaffold acts as a container to encapsulate chemotherapeutic drug doxorubicin. In vitro and in vivo studies verify that the catalase-like nanohybrid significantly enhances the radiotherapy effect, alleviating tumor hypoxia and achieving synergistic anticancer efficacy. This hybrid nanomaterial remarkably suppresses the tumor growth with minimized systemic toxicity, opening new horizons for the next generation of theranostic nanomedicines.


Subject(s)
Catalase/chemistry , Chemoradiotherapy/methods , Metal-Organic Frameworks/chemistry , Humans
13.
Angew Chem Int Ed Engl ; 58(26): 8799-8803, 2019 06 24.
Article in English | MEDLINE | ID: mdl-31034679

ABSTRACT

Single molecular nanoparticles (SMNPs) integrating imaging and therapeutic capabilities exhibit unparalleled advantages in cancer theranostics, ranging from excellent biocompatibility, high stability, prolonged blood lifetime to abundant tumor accumulation. Herein, we synthesize a sophisticated porphyrin nanocage that is further functionalized with twelve polyethylene glycol arms to prepare SMNPs (porSMNPs). The porphyrin nanocage embedded in porSMNPs can be utilized as a theranostic platform. PET imaging allows dynamic observation of the bio-distribution of porSMNPs, confirming their excellent circulation time and preferential accumulation at the tumor site, which is attributed to the enhanced permeability and retention effect. Moreover, the cage structure significantly promotes the photosensitizing effect of porSMNs by inhibiting the π-π stacking interactions of the photosensitizers, ablating of the tumors without relapse by taking advantage of photodynamic therapy.


Subject(s)
Nanoparticles/chemistry , Nanotechnology/methods , Porphyrins/chemistry , Theranostic Nanomedicine/methods , Humans
14.
J Am Chem Soc ; 140(13): 4666-4677, 2018 04 04.
Article in English | MEDLINE | ID: mdl-29543442

ABSTRACT

This article describes the fabrication of nanosized magneto-vesicles (MVs) comprising tunable layers of densely packed superparamagnetic iron oxide nanoparticles (SPIONs) in membranes via cooperative assembly of polymer-tethered SPIONs and free poly(styrene)- b-poly(acrylic acid) (PS- b-PAA). The membrane thickness of MVs could be well controlled from 9.8 to 93.2 nm by varying the weight ratio of PS- b-PAA to SPIONs. The increase in membrane thickness was accompanied by the transition from monolayer MVs, to double-layered MVs and to multilayered MVs (MuMVs). This can be attributed to the variation in the hydrophobic/hydrophilic balance of polymer-grafted SPIONs upon the insertion and binding of PS- b-PAA onto the surface of nanoparticles. Therapeutic agents can be efficiently encapsulated in the hollow cavity of MVs and the release of payload can be tuned by varying the membrane thickness of nanovesicles. Due to the high packing density of SPIONs, the MuMVs showed the highest magnetization and transverse relaxivity rate ( r2) in magnetic resonance imaging (MRI) among these MVs and individual SPIONs. Upon intravenous injection, doxorubicin-loaded MuMVs conjugated with RGD peptides could be effectively enriched at tumor sites due to synergetic effect of magnetic and active targeting. As a result, they exhibited drastically enhanced signal in MRI, improved tumor delivery efficiency of drugs as well as enhanced antitumor efficacy, compared with groups with only magnetic or active targeting strategy. The unique nanoplatform may find applications in effective disease control by delivering imaging and therapy to organs/tissues that are not readily accessible by conventional delivery vehicles.


Subject(s)
Drug Delivery Systems , Magnetics , Magnetite Nanoparticles/chemistry , Magnetic Resonance Imaging , Permeability
15.
J Am Chem Soc ; 140(25): 8005-8019, 2018 06 27.
Article in English | MEDLINE | ID: mdl-29874067

ABSTRACT

Nanomedicines have achieved several breakthroughs in cancer treatment over the past decades; however, their potential immunotoxicities are ignored, which results in serious adverse effects and greatly reduces the potential in clinical translation. Herein, we innovatively develop a theranostic supramolecular polymer using ß-cyclodextrin as the host and camptothecin (CPT) as the guest linked by a glutathione-cleavable disulfide bond. The supramolecular polymerization remarkably increases the solubility of CPT by a factor of 232 and effectively inhibits its lactone ring opening in physiological environment, which is favorable for intravenous formulation and maintenance of the therapeutic efficacy. Supramolecular nanoparticles can be prepared through orthogonal self-assembly driven by π-π stacking interaction, host-guest complexation, and hydrogen bonds. The sophisticated nanomedicine constructed from the obtained supramolecular polymer can be specifically delivered to tumor sites and rapidly excreted from body after drug release, thus effectively avoiding systemic toxicity, especially long-term immunotoxicity. In vivo investigations demonstrate this supramolecular nanomedicine possesses superior antitumor performance and antimetastasis capability. This pioneering example integrating the advantages of the dynamic nature of supramolecular chemistry and nanotechnology provides a promising platform for cancer theranostics.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Camptothecin/administration & dosage , Glutathione/chemistry , beta-Cyclodextrins/chemistry , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/therapeutic use , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Camptothecin/adverse effects , Camptothecin/chemistry , Camptothecin/therapeutic use , Female , HeLa Cells , Humans , Nanomedicine , Neoplasms/diagnostic imaging , Neoplasms/drug therapy , Positron-Emission Tomography , Solubility
16.
Bioconjug Chem ; 29(9): 3213-3221, 2018 09 19.
Article in English | MEDLINE | ID: mdl-30105912

ABSTRACT

Prostate cancer is the most frequently diagnosed malignant tumor in men worldwide. Prostate-specific membrane antigen (PSMA) is a surface molecule specifically expressed by prostate tumors that has been shown to be a valid target for internal radionuclide therapy in both preclinical and clinical settings. The most common radiotherapeutic agent is the small molecule 177Lu-PSMA-617, which is under clinical evaluation in multiple countries. Nevertheless, its efficacy in causing tumor regression is still suboptimal, even when administered in several cycles per patient, perhaps due to poor pharmacokinetics (PK), which limits uptake by the tumor cells. We postulated that the addition of the Evans blue (EB) moiety to PSMA-617 would improve the PK by extending circulation half-life, which would increase tumor uptake and improve radiotherapeutic efficacy. PSMA-617 was modified by conjugation of a 2-thiol acetate group onto the primary amine and thereafter reacted with a maleimide functional group of an EB derivative, to give EB-PSMA-617. The PK and radiotherapeutic efficacy of 90Y- or 177Lu-EB-PSMA-617 was compared to the clinically used radiopharmaceutical 90Y- or 177Lu- PSMA-617 in PC3-PIP tumor-bearing mice. EB-PSMA-617 retained binding to serum albumin as well as a high internalization rate by tumor cells. Upon injection, metal-labeled EB-PSMA-617 demonstrated an extended blood half-life compared to PSMA-617 and, thereby, prolonged the time window for binding to PSMA. The improved PK of EB-PSMA-617 resulted in significantly higher accumulation in PSMA+ tumors and highly effective radiotherapeutic efficacy. Remarkably, a single dose of 1.85 MBq of 90Y- or 177Lu-EB-PSMA-617 was sufficient to eradicate established PMSA+ tumors in mice. No significant body weight loss was observed, suggesting little to no gross toxicity. The construct described here, EB-PSMA-617, may improve the radiotherapeutic efficacy for patients with PSMA-positive tumors by reducing both the amount of activity needed for therapy as well as the frequency of administration, as compared to PSMA-617.


Subject(s)
Dipeptides/therapeutic use , Evans Blue/administration & dosage , Heterocyclic Compounds, 1-Ring/therapeutic use , Lutetium/therapeutic use , Prostatic Neoplasms/drug therapy , Radiopharmaceuticals/therapeutic use , Yttrium Radioisotopes/therapeutic use , Animals , Dipeptides/chemistry , Dipeptides/pharmacokinetics , Heterocyclic Compounds, 1-Ring/chemistry , Heterocyclic Compounds, 1-Ring/pharmacokinetics , Humans , Lutetium/chemistry , Lutetium/pharmacokinetics , Male , Mice , Positron-Emission Tomography , Prostate-Specific Antigen , Radiopharmaceuticals/chemistry , Radiopharmaceuticals/pharmacokinetics , Xenograft Model Antitumor Assays , Yttrium Radioisotopes/chemistry , Yttrium Radioisotopes/pharmacokinetics
17.
Bioconjug Chem ; 29(7): 2309-2315, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29865797

ABSTRACT

Several radioligands targeting prostate-specific membrane antigen (PSMA) have been clinically introduced as a new class of radiotheranostics for the treatment of prostate cancer. Among them, ((( R)-1-carboxy-2-mcercaptoethyl)carbamoyl)-l-glutamic acid (MCG) has been successfully labeled with radioisotopes for prostate cancer imaging. The aim of this study is to conjugate MCG with an albumin binding moiety to further improve the in vivo pharmacokinetics. MCG was conjugated with an Evans blue (EB) derivative for albumin binding and a DOTA chelator. PSMA positive (PC3-PIP) and PSMA negative (PC3) cells were used for both in vitro and in vivo studies. Longitudinal PET imaging was performed at 1, 4, 24, and 48 h post-injection to evaluate the biodistribution and tumor uptake of 86Y-DOTA-EB-MCG. DOTA-EB-MCG was also labeled with 90Y for radionuclide therapy. Besides tumor growth measurement, tumor response to escalating therapeutic doses were also evaluated by immunohistochemistry and fluorescence microscopy. Based on quantification from 86Y-DOTA-EB-MCG PET images, the tracer uptake in PC3-PIP tumors increased from 22.33 ± 2.39%ID/g at 1 h post-injection (p.i.), to the peak of 40.40 ± 4.79%ID/g at 24 h p.i. Administration of 7.4 MBq of 90Y-DOTA-EB-MCG resulted in significant regression of tumor growth in PSMA positive xenografts. No apparent toxicity or body weight loss was observed in all treated mice. Modification of MCG with an Evans blue derivative resulted in a highly efficient prostate cancer targeting agent (EB-MCG), which showed great potential in prostate cancer treatment after being labeled with therapeutic radioisotopes.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Glutamates/chemistry , Prostatic Neoplasms/diagnostic imaging , Prostatic Neoplasms/radiotherapy , Animals , Evans Blue/chemistry , Heterocyclic Compounds/chemistry , Heterografts , Humans , Male , Mice , Organometallic Compounds/chemistry , PC-3 Cells , Positron-Emission Tomography , Yttrium Isotopes
18.
Bioconjug Chem ; 29(2): 410-419, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29254329

ABSTRACT

Radiolabeled bombesin (BBN) analogs have long been used for developing gastrin-releasing peptide receptor (GRPR) targeted imaging probes, and tracers with excellent in vivo performance including high tumor uptake, high contrast, and favorable pharmacokinetics are highly desired. In this study, we compared the 68Ga-labeled GRPR agonist (Gln-Trp-Ala-Val-Gly-His-Leu-Met-NH2, BBN7-14) and antagonist (d-Phe-Gln-Trp-Ala-Val-Gly-His-Sta-Leu-NH2, RM26) for the positron emission tomography (PET) imaging of prostate cancer. The in vitro stabilities, receptor binding, cell uptake, internalization, and efflux properties of the probes 68Ga-1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA)-Aca-BBN7-14 and 68Ga-NOTA-poly(ethylene glycol)3 (PEG3)-RM26 were studied in PC-3 cells, and the in vivo GRPR targeting abilities and kinetics were investigated using PC-3 tumor xenografted mice. BBN7-14, PEG3-RM26, NOTA-Aca-BBN7-14, and NOTA-PEG3-RM26 showed similar binding affinity to GRPR. In PC-3 tumor-bearing mice, the tumor uptake of 68Ga-NOTA-PEG3-RM26 remained at around 3.00 percentage of injected dose per gram of tissue within 1 h after injection, in contrast with 68Ga-NOTA-Aca-BBN7-14, which demonstrated rapid elimination and high background signal. Additionally, the majority of the 68Ga-NOTA-PEG3-RM26 remained intact in mouse serum at 5 min after injection, while almost all of the 68Ga-NOTA-Aca-BBN7-14 was degraded under the same conditions, demonstrating more-favorable in vivo pharmacokinetic properties and metabolic stabilities of the antagonist probe relative to its agonist counterpart. Overall, the antagonistic GRPR targeted probe 68Ga-NOTA-PEG3-RM26 is a more-promising candidate than the agonist 68Ga-NOTA-Aca-BBN7-14 for the PET imaging of prostate cancer patients.


Subject(s)
Gallium Radioisotopes/chemistry , Peptides/chemistry , Positron-Emission Tomography/methods , Prostate/diagnostic imaging , Prostatic Neoplasms/diagnostic imaging , Receptors, Bombesin/agonists , Receptors, Bombesin/antagonists & inhibitors , Amino Acid Sequence , Animals , Cell Line, Tumor , Female , Gallium Radioisotopes/pharmacokinetics , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/pharmacokinetics , Heterocyclic Compounds, 1-Ring , Humans , Male , Mice , Mice, Inbred BALB C , Peptides/pharmacokinetics
19.
Proc Natl Acad Sci U S A ; 112(45): E6129-38, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26504197

ABSTRACT

It is a critically important challenge to rapidly design effective vaccines to reduce the morbidity and mortality of unexpected pandemics. Inspired from the way that most enveloped viruses hijack a host cell membrane and subsequently release by a budding process that requires cell membrane scission, we genetically engineered viral antigen to harbor into cell membrane, then form uniform spherical virus-mimetic nanovesicles (VMVs) that resemble natural virus in size, shape, and specific immunogenicity with the help of surfactants. Incubation of major cell membrane vesicles with surfactants generates a large amount of nano-sized uniform VMVs displaying the native conformational epitopes. With the diverse display of epitopes and viral envelope glycoproteins that can be functionally anchored onto VMVs, we demonstrate VMVs to be straightforward, robust and tunable nanobiotechnology platforms for fabricating antigen delivery systems against a wide range of enveloped viruses.


Subject(s)
Antigens/metabolism , Drug Delivery Systems/methods , Nanostructures/chemistry , Transport Vesicles/chemistry , Viral Vaccines/metabolism , Nanotechnology/methods , Nanotechnology/trends , Phospholipids/analysis , Recombinant Proteins/metabolism , Surface-Active Agents/analysis
20.
Angew Chem Int Ed Engl ; 57(24): 7066-7070, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29624828

ABSTRACT

Polymeric micelle-based drug delivery systems have dramatically improved the delivery of small molecular drugs, yet multiple challenges remain to be overcome. A polymeric nanomedicine has now been engineered that possesses an ultrahigh loading (59 %) of a glutathione (GSH)-sensitive heterodimeric multifunctional prodrug (HDMP) to effectively co-deliver two synergistic drugs to tumors. An HDMP comprising of chemotherapeutic camptothecin (CPT) and photosensitizer 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-α (HPPH) was conjugated via a GSH-cleavable linkage. The intrinsic fluorogenicity and label-free radio-chelation (64 Cu) of HPPH enabled direct drug monitoring by fluorescence imaging and positron emission tomography (PET). Through quantitative PET imaging, HDMP significantly improves drug delivery to tumors. The high synergistic therapeutic efficacy of HDMP-loaded NPs highlights the rational design of HDMP, and presents exciting opportunities for polymer NP-based drug delivery.


Subject(s)
Antineoplastic Agents/administration & dosage , Camptothecin/administration & dosage , Delayed-Action Preparations/metabolism , Glutathione/metabolism , Photosensitizing Agents/administration & dosage , Prodrugs/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Camptothecin/therapeutic use , Cell Line, Tumor , Delayed-Action Preparations/chemistry , Drug Monitoring , Drug Synergism , HCT116 Cells , Humans , Mice , Neoplasms/drug therapy , Neoplasms/metabolism , Photosensitizing Agents/therapeutic use , Polymers/chemistry , Polymers/metabolism , Prodrugs/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL