Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Control Release ; 370: 95-109, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38642859

ABSTRACT

Sonodynamic therapy (SDT) has emerged as a useful approach for tumor treatment. However, its widespread application is impeded by poor pharmacokinetics of existing sonosensitizers. Here we developed a metal-organic nanoplatform, wherein a small-molecule sonosensitizer (hematoporphyrin monomethyl ether, HMME) was ingeniously coordinated with zirconium, resulting in a multifunctional nanosonosensitizer termed Zr-HMME. Through post-synthetic modifications involving PEGylation and tumor-targeting peptide (F3) linkage, a nanoplatform capable of homing on melanoma was produced, which could elicit robust immune responses to suppress tumor lung metastasis in the host organism. Importantly, after seamless incorporation of positron-emitting 89Zr into this nanosonosensitizer, positron emission tomography (PET) could be used to monitor its in vivo pharmacokinetics. PET imaging studies revealed that this nanoplatform exhibited potent tumor accumulation and strong in vivo stability. Using intrinsic fluorescence from HMME, a dual-modal diagnostic capability (fluorescence and PET) was confirmed for this nanosonosensitizer. In addition, the mechanisms of how this nanoplatform interacted with immune system were also investigated. The collective data proved that the coordination structure between small-molecule drug cargos and metals may enhance the functions of each other while mitigating their weaknesses. This straightforward approach can expand the potential applications of suitable drug molecules.


Subject(s)
Hematoporphyrins , Positron-Emission Tomography , Zirconium , Zirconium/chemistry , Zirconium/pharmacokinetics , Animals , Positron-Emission Tomography/methods , Cell Line, Tumor , Hematoporphyrins/administration & dosage , Hematoporphyrins/chemistry , Hematoporphyrins/pharmacokinetics , Melanoma/diagnostic imaging , Melanoma/drug therapy , Mice, Inbred C57BL , Ultrasonic Therapy/methods , Mice , Melanoma, Experimental/therapy , Melanoma, Experimental/diagnostic imaging , Nanoparticles/chemistry , Female , Radioisotopes/administration & dosage
2.
Acta Biomater ; 174: 372-385, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38072226

ABSTRACT

Targeted delivery of therapeutic drugs to fibrosis-promoting macrophages (FPMs) holds promise as a challenging yet effective approach for the treatment of idiopathic pulmonary fibrosis (IPF). Here, nanocarriers composed of Mn-curcumin metal-organic frameworks (MOFs) were utilized to deliver the immune inhibitor BLZ-945 to the lungs, with the goal of depleting fibrosis-promoting macrophages (FPMs) from fibrotic lung tissues. FPM targeting was achieved by functionalizing the nanocarrier surface with an M2-like FPM binding peptide (M2pep). As a result, significant therapeutic benefits were observed through the successful depletion of approximately 80 % of the M2-like macrophages (FPMs) in a bleomycin-induced fibrosis mouse model treated with the designed M2-like FPM-targeting nanoparticle (referred to as M2NP-BLZ@Mn-Cur). Importantly, the released Mn2+ and curcumin after the degradation of M2NP-BLZ@Mn-Cur accumulated in the fibrotic lung tissue, which can alleviate inflammation and oxidative stress reactions, thereby further improving IPF therapy. This study presents a novel strategy with promising prospects for molecular-targeted fibrosis therapy. STATEMENT OF SIGNIFICANCE: Metal-organic frameworks (MOFs)- based nanocarriers equipped with both fibrosis-promoting macrophage (FPM)-specific targeting ability and therapeutic drugs are appealing for pulmonary fibrosis treatment. Here, we prepared M2pep (an M2-like FPM binding peptide)-modified and BLZ945 (a small molecule inhibitor of CSF1/CSF-1R axis)-loaded Mn-curcumin MOF nanoparticles (M2NP-BLZ@Mn-Cur) for pulmonary fibrosis therapy. The functionalized M2NP-BLZ@Mn-Cur nanoparticles can be preferentially taken up by FPMs, resulting in their depletion from fibrotic lung tissues. In addition, Mn2+and curcumin released from the nanocarriers have anti-inflammation and immune regulation effects, which further enhance the antifibrotic effect of the nanoparticles.


Subject(s)
Curcumin , Idiopathic Pulmonary Fibrosis , Metal-Organic Frameworks , Mice , Animals , Metal-Organic Frameworks/pharmacology , Curcumin/pharmacology , Curcumin/therapeutic use , Curcumin/chemistry , Macrophages/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Peptides/pharmacology
3.
ACS Appl Mater Interfaces ; 14(51): 56510-56524, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36516041

ABSTRACT

Shikonin (Shik), a natural pigment, has received growing interest in various biomedical fields due to its anti-inflammatory, antitumor, antimicrobial, and antioxidant ability. However, some inherent characteristics of Shik, such as its virulence, low bioavailability, and poor solubility, have limited its biomedical applicability. Here, we reported a facile synthetic method to produce the Shik-iron (III) nanoparticles (Shik-Fe NPs), which could overcome these limitations of Shik. The synthesized Shik-Fe NPs possessed a uniform size range of 110 ± 10 nm, negative surface charges, good water dispersity, and high safety. Iron distributed uniformly inside Shik-Fe NPs, and iron constituted 20% of total mass in PEGylated Shik-Fe NPs. When interacting with activated macrophages, Shik-Fe NPs significantly reduced the level of cellular inflammatory factors, for example, iNOS, IL-1ß, and TNF-α. Furthermore, the Shik-Fe NPs demonstrated synergistic anti-inflammation and anti-bacterial properties in vivo, since they could release Fe3+ and Shik to eradicate bacteria (Staphylococcus aureus and P. aeruginosa were used as model microbes here) during wound infections and provide full recovery for scald wounds. Collectively, the study established a dual-functional Shik-derived nanoplatform, which could be useful for the treatment of various inflammation-involved diseases.


Subject(s)
Anti-Infective Agents , Nanoparticles , Ferric Compounds , Staphylococcus aureus , Anti-Inflammatory Agents , Iron , Anti-Bacterial Agents/pharmacology
4.
J Nanobiotechnology ; 20(1): 494, 2022 Nov 24.
Article in English | MEDLINE | ID: mdl-36424645

ABSTRACT

BACKGROUND: Carbon monoxide (CO) is an important signaling molecule participating in multiple biological functions. Previous studies have confirmed the valuable roles of CO in cancer therapies. If the CO concentration and distribution can be controlled in tumors, new cancer therapeutic strategy may be developed to benefit the patient survival. RESULTS: In this study, a UiO-67 type metal-organic framework (MOF) nanoplatform was produced with cobalt and ruthenium ions incorporated into its structure (Co/Ru-UiO-67). Co/Ru-UiO-67 had a size range of 70-90 nm and maintained the porous structure, with cobalt and ruthenium distributed uniformly inside. Co/Ru-UiO-67 was able to catalyze carbon dioxide into CO upon light irradiation in an efficient manner with a catalysis speed of 5.6 nmol/min per 1 mg Co/Ru-UiO-67. Due to abnormal metabolic properties of tumor cells, tumor microenvironment usually contains abundant amount of CO2. Co/Ru-UiO-67 can transform tumor CO2 into CO at both cellular level and living tissues, which consequently interacts with relevant signaling pathways (e.g. Notch-1, MMPs etc.) to adjust tumor microenvironment. With proper PEGylation (pyrene-polyacrylic acid-polyethylene glycol, Py-PAA-PEG) and attachment of a tumor-homing peptide (F3), functionalized Co/Ru-UiO-67 could accumulate strongly in triple-negative MDA-MB-231 breast tumors, witnessed by positron emission tomography (PET) imaging after the addition of radioactive zirconium-89 (89Zr) into Co-UiO-67. When applied in vivo, Co/Ru-UiO-67 could alter the local hypoxic condition of MDA-MB-231 tumors, and work synergistically with tirapazamine (TPZ). CONCLUSION: This nanoscale UiO-67 MOF platform can further our understanding of CO functions while produce CO in a controllable manner during cancer therapeutic administration.


Subject(s)
Metal-Organic Frameworks , Ruthenium , Triple Negative Breast Neoplasms , Humans , Metal-Organic Frameworks/pharmacology , Metal-Organic Frameworks/chemistry , Carbon Monoxide , Ruthenium/pharmacology , Ruthenium/chemistry , Triple Negative Breast Neoplasms/drug therapy , Carbon Dioxide , Cobalt , Tumor Microenvironment
5.
Adv Healthc Mater ; 11(8): e2102270, 2022 04.
Article in English | MEDLINE | ID: mdl-35032116

ABSTRACT

Formation of protein corona on nanomaterials surface in vivo is usually considered as an unpredictable event for a predefined targeted delivery system for malignant cancers. In most situations, these protein coronas substantially change targeting efficiency or even cause adverse reactions which both hinder the clinical translation of the cargo-delivery systems. Active customization of protein corona onto nanomaterials surfaces can benefit their biomedical performances and open up new opportunities in construction of targeted delivery systems. Herein, lipid-PEG/pheophytin carbon dots (LPCDs) are prepared from natural chlorophyll and integrate seamlessly with positron emission tomography imaging, near-infrared fluorescence imaging, and photodynamic therapy capacity. In vitro measurements demonstrate that the LPCDs can actively absorb apolipoproteins into the protein corona to enhance their uptakes in breast cancer cells. In vivo studies confirm that LPCDs can give accurate delineation of metastatic breast cancer foci from surrounding normal tissues with multimodal biomedical functions. The feasibility of using LPCDs as a multimodal imaging and cancer-targeting nanoplatform may provide impetus for developing precise yet facile protein corona-targeted delivery systems for future clinical practice.


Subject(s)
Breast Neoplasms , Carbon , Nanoparticles , Photochemotherapy , Protein Corona , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Carbon/administration & dosage , Carbon/chemistry , Female , Humans , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Pheophytins/therapeutic use , Protein Corona/metabolism
6.
Bioconjug Chem ; 32(10): 2195-2204, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34591471

ABSTRACT

Nanoscale metal-organic frameworks (nMOFs) are a unique type of hybrid materials, which are broadly applicable as cargo delivery systems. However, the relatively low material stability and insufficient cancer cell interacting capacity have limited nMOFs' applications in cancer theranostics. Herein, a zirconium-based nMOF UiO-66-N3 was synthesized, and its surface was covalently functionalized with alkyne-containing polyethylene glycol (PEG) via the azide-alkyne click chemistry. After that, F3 peptide was attached for targeting of cancer cells (the material was denoted as UiO-66-PEG-F3). Doxorubicin (DOX) served as a therapeutic drug and a fluorescent label in this study, and it was transported into UiO-66-PEG conjugates with sufficient drug loading efficiency. pH-responsive release of DOX from UiO-66 conjugates was witnessed. The structural integrity of UiO-66-N3 was maintained post the surface modification process. Flow cytometry and confocal fluorescence microscopy revealed that DOX/UiO-66-PEG-F3 had stronger accumulation in MDA-MB-231 cells (nucleolin+) compared with DOX/UiO-66-PEG. In order to track the pharmacokinetic behavior (organ distribution profile) in vivo, the positron-emitting zirconium-89 (89Zr) was incorporated into UiO-66-N3. Similar PEGylation and F3 peptide conjugation resulted in the formation of 89Zr-UiO-66-PEG-F3. Serial positron emission tomography (PET) imaging demonstrated that the preferential accumulation of 89Zr-UiO-66-PEG-F3 in MDA-MB-231 tumors, and their liver clearance was faster than PEGylated UiO-66 using noncovalent methods. Thus, the PEGylated nMOFs using covalent strategies may find broad application in future cancer theranostics.


Subject(s)
Drug Carriers , Metal-Organic Frameworks
7.
Mol Pharm ; 18(4): 1690-1698, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33734721

ABSTRACT

The urokinase plasminogen activator (uPA) and its cofactors are important regulators of tumor initiation and progression (including metastasis), and its overexpression is associated with unfavorable situations in cancer patients. We have previously used positron emission tomography (PET) imaging with a radiolabeled monoclonal antibody against the uPA (named ATN-291) to detect the uPA signaling activity in various cancer types; however, good tumor contrast can only be observed 24 h postinjection. To shorten the antibody circulation time and decrease interactions of ATN-291 with the mononuclear phagocyte system (MPS), our goal in this study is to develop an engineered antibody fragment (F(ab')2) from the parent antibody. By pepsin digestion and chromatography purification, ATN-291 F(ab')2 was obtained and characterized. Subsequently, it was conjugated with NOTA-Bn-NCS or fluorescein isothiocyanate (FITC) for PET imaging and fluorescence-mediated cellular analysis (i.e., flow cytometry or fluorescence microscopy). We confirmed that ATN-291 F(ab')2 still maintained a good targeting efficacy for the uPA in MDA-MB-231 cells (uPA+) and it had a faster blood clearance speed compared with ATN-291, while its interaction with MPS has been significantly decreased. In rodent tumor xenografts, radiolabeled ATN-291 F(ab')2 had a selective and persistent uptake in MDA-MB-231 tumors, with an early tumor-to-blood ratio of 1.3 ± 0.8 (n = 4) at 2 h postinjection from PET imaging. During our observation, radiolabeled ATN-291 F(ab')2 was excreted from both renal and hepatobiliary pathways. Radiolabeled ATN-291 F(ab')2 was also used for detecting uPA fluctuation during the tumor treatment in test animals. We concluded that radiolabeled ATN-291 F(ab')2 could be used as fast as PET cancer diagnostics with versatile applicability.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Immunoglobulin Fab Fragments/administration & dosage , Membrane Proteins/antagonists & inhibitors , Positron-Emission Tomography/methods , Triple Negative Breast Neoplasms/diagnosis , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/genetics , Female , Fluorescein-5-isothiocyanate/chemistry , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Membrane Proteins/metabolism , Mice , Protein Engineering , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...