Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Am J Dermatopathol ; 46(10): 693-695, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38916239

ABSTRACT

ABSTRACT: Extramammary Paget disease (EMPD) is a rare cutaneous malignancy, typically presenting as eczema-like lesions in areas rich in apocrine glands such as the perineum. Here, we report a case of EMPD presenting as a prominent pedunculated neoplasm in a 65-year-old woman. Despite initial misdiagnosis and treatment, biopsy confirmed EMPD infiltration. Following surgical excision, the patient developed brain metastases, indicating a poor prognosis. EMPD's pathogenesis remains unclear, but distinguishing primary from secondary forms is crucial for prognosis and treatment. Our case underscores the importance of recognizing atypical EMPD presentations for timely intervention and improved outcomes.


Subject(s)
Paget Disease, Extramammary , Vulvar Neoplasms , Humans , Female , Aged , Vulvar Neoplasms/pathology , Paget Disease, Extramammary/pathology , Paget Disease, Extramammary/surgery , Paget Disease, Extramammary/diagnosis , Biopsy , Brain Neoplasms/pathology , Fatal Outcome
2.
Adv Healthc Mater ; 12(8): e2202418, 2023 03.
Article in English | MEDLINE | ID: mdl-36459700

ABSTRACT

The hypoxic character of tumors and the poor targeting ability of photosensitizers often limit the efficacy of photodynamic therapy (PDT). In recent years, the discovery of metal nanoenzymes and nanocarriers has improved PDT. Thereby, to improve the effective utilization of photosensitizers and oxygen (O2 ) in tumors, herein, a nanosystem (LS-HB@HvCeO2 -NRP1 mAb, LHCN1) is reported, in which a hollow virus-like cerium oxide (HvCeO2 ) is surface-decorated with tumor-targeting neuropilin-1 monoclonal antibody (NRP1 mAb), and loaded with a photosensitizer (chlorin e6-C-15-ethyl ester, LS-HB). In vitro and in vivo experiments demonstrate that LHCN1 can efficiently accumulate within the tumor sites via the targeting guidance of NRP1 mAb and is then rapidly endocytosed into cells. Furthermore, HvCeO2 with catalase-mimetic activity can decompose the endogenous hydrogen peroxide (H2 O2 ) to promote O2 via the valence transformation between Ce4+ and Ce3+ , relieving tumor hypoxia and improving the PDT efficacy. Upon near-infrared laser irradiation, LS-HB produces large amounts of cytotoxic reactive oxygen species. Moreover, LHCN1 is used in fluorescence/photoacoustic multimodal imaging for in vivo drug localization, and its use in PDT evidently helps inhibit tumor growth with no apparent toxicity to normal tissues. Thus, LHCN1 may provide a promising strategy for precise tumor-specific diagnosis and treatment.


Subject(s)
Nanoparticles , Photochemotherapy , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Oncogenic Viruses , Cell Line, Tumor , Hydrogen Peroxide , Oxygen
3.
Adv Healthc Mater ; 11(16): e2102770, 2022 08.
Article in English | MEDLINE | ID: mdl-35575205

ABSTRACT

The self-delivery of sonosensitizers and immunomodulators to tumor areas, which is highly recommended for enhancing sonodynamic immunotherapy, remains a challenge. Herein, a self-delivering nanodrug (HB-NLG8189, drug loading: ≈100 wt%) is developed by the small-molecule self-assembly of "HB" (a new clinical photosensitizer) and NLG8189 (indoleamine-(2,3)-dioxygenase (IDO) pathway inhibitor) for sonodynamic-augmented immunotherapy; this preparation method ensures the absence of excipient-related toxicity and immunogenicity. To evade immune recognition and prolong the circulation time, the HB-NLG8189 nanodrugs are camouflaged using macrophage cell membranes (MPCMs). The constructed HB-NLG8189@MPCM nanodrugs show an ability to preferentially accumulate within tumors. Upon ultrasound triggering, the HB-NLG8189@MPCM is able to generate reactive oxygen species efficiently for robust sonodynamic therapy; it induces immunogenic cell death, initiates an antitumor immune response to activate tumor-specific effector T cells, and promotes the secretion of inflammatory cytokines. The concomitant delivery of NLG8189 reverses the immunosuppressive tumor microenvironment by restraining IDO-1 activation and the intratumoral infiltration of regulatory T cells. Sonodynamic-augmented immunotherapy with HB-NLG8189@MPCM significantly inhibits the growth of both primary and distant tumors with little systemic toxicity. The biomimetic self-delivery nanodrug provides a promising paradigm for improving sonodynamic immunotherapy.


Subject(s)
Nanoparticles , Neoplasms , Cell Line, Tumor , Humans , Immunologic Factors/therapeutic use , Immunotherapy , Indoleamine-Pyrrole 2,3,-Dioxygenase , Macrophages/pathology , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Neoplasms/pathology , Tumor Microenvironment
4.
Adv Healthc Mater ; 11(6): e2102060, 2022 03.
Article in English | MEDLINE | ID: mdl-34894092

ABSTRACT

The anti-tumor efficacy of single photodynamic therapy (PDT) and radiotherapy (RT) has been greatly affected by inadequate tumor uptake of photo/radiation sensitizers, limited laser penetration depth, and radiation sickness caused by high doses of X-rays. Here, the authors report a biomimetic coronavirus-inspired hollow mesoporous gadolinium/bismuth nanocarrier loaded with a new NIR photosensitizer HB (termed as HB@VHMBi-Gd) for magnetic resonance imaging (MRI)-guided synergistic photodynamic-RT. HB@VHMBi-Gd displayed a faster cellular uptake rate than the conventional spherical HMBi-Gd loaded with HB (HB@SHMBi-Gd) because of rough surface-enhanced adhesion. After intravenous injection, HB@VHMBi-Gd is efficiently delivered to the tumor and rapidly invades the tumor cells by surface spikes. Interestingly, lysosomal acidity can trigger the degradation of VHMBi-Gd to produce ultrasmall nanoparticles to amplify the X-ray attenuation ability and enhance MRI contrast and radiosensitization. Under laser and X-ray irradiation, HB@VHMBi-Gd significantly enhances 1 O2 generation from HB to induce activation of caspase 9/3 and inhibition of C-myc, while enhancing hydroxyl radical generation from Bi2 O3 to induce intense DNA breakage. By synergistically inducing cell apoptosis by distinct reactive oxygen species (ROS), HB@VHMBi-Gd exhibits superior anticancer efficacy with ≈90% tumor inhibition. They envision that biomimetic virus-inspired hollow hybrid metal nanoparticles can provide a promising strategy for imaging-guided synergistic photodynamic-RT.


Subject(s)
Metal Nanoparticles , Nanoparticles , Photochemotherapy , Bismuth , Cell Line, Tumor , Gadolinium , Magnetic Resonance Imaging , Photochemotherapy/methods , Theranostic Nanomedicine/methods
5.
Front Oncol ; 11: 657008, 2021.
Article in English | MEDLINE | ID: mdl-34336654

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive malignancy with extremely limited treatment; the effective targeting strategy stays an urgent unmet need. Neuropilin-2 (NRP2), a multifunctional transmembrane non-tyrosine-kinase glycoprotein, enhances various signal transduction pathways to modulate cancer progression. However, the application value of NRP2 as a therapeutic target in pancreatic cancer is still unclear. Here, we detected the elevated NRP2 was associated with the poor prognosis of pancreas carcinoma. The mouse monoclonal antibody targeting NRP2 (N2E4) that could specifically bind to PDAC cells was developed. Moreover, N2E4 inhibits PDAC proliferation, migration, and invasion in vitro, and repressed growth and metastasis in vivo. Mechanistically, the effect of N2E4 was mainly related to the blocking of interaction between NRP2 with integrinß1 to inhibit FAK/Erk/HIF-1a/VEGF signaling. Therefore, N2E4 has the potential for targeting therapy of PDAC. This study lays a foundation for the future development of NRP2-based targeted therapy for PDAC.

6.
Drug Deliv ; 28(1): 1769-1784, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34470548

ABSTRACT

Traditional combined photodynamic and photothermal therapy (PDT/PTT) was limited in clinical treatment of cancer due to the exceptionally low drug delivery efficiency to tumor sites and the activation by laser excitation with different wavelengths. We have accidentally discovered that our synthesized chlorin e6-C-15-ethyl ester (HB, a new type of photosensitizer) be activated by a laser with an excitation wavelength of 660 nm. Herein, we utilized Au nanorods (AuNRs) as 660 nm-activated PTT carriers to be successively surface-functionalized with HB and tumor-targeting peptide cyclic RGD (cRGD) to develop HB-AuNRs@cRGD for single NIR laser-induced targeted PDT/PTT. The HB-AuNRs@cRGD could be preferentially accumulated within tumor sites and rapidly internalized by cancer cells. Thereby, the HB-AuNRs@cRGD could exhibit amplified therapeutic effects by producing both significant reactive oxygen species (ROS) and hyperthermia simultaneously under the guidance of fluorescence imaging. The tumor inhibition rate on ECA109 esophageal cancer model was approximately 77.04%, and the negligible systematic toxicity was observed. This study proposed that HB-AuNRs@cRGD might be a promising strategy for single NIR laser-induced and imaging-guided targeted bimodal phototherapy.


Subject(s)
Gold/chemistry , Nanotubes/chemistry , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photothermal Therapy/methods , Animals , Cell Line , Female , Low-Level Light Therapy , Mice , Mice, Inbred BALB C , Photosensitizing Agents/administration & dosage , Photosensitizing Agents/pharmacokinetics , Reactive Oxygen Species/metabolism
7.
Nanoscale ; 13(1): 185-194, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33325961

ABSTRACT

It is interesting yet challenging to design theranostic nanoplatforms for the accurate diagnosis and therapy of diseases; these nanoplatforms consist of single contrast-enhanced imaging or therapeutic agents, and they possess their own unique shortcomings that limit their widespread bio-medical applications. Therefore, designing a potential theranostic agent is an emerging approach for the synergistic diagnosis and therapeutics in bio-medical applications. Herein, a lanthanide-loaded (NaLnF4) heterostructure BiOCl ultrathin nanosheet (BiNS@NaLnF4) as a theranostic agent was synthesized facilely by a solvothermal protocol. BiNS@NaLnF4 was employed as a multi-modal contrast agent for computed tomography (CT) and magnetic resonance imaging (MRI), showing a high-performance X-ray absorption contrast effect, an outstanding T1-weighted imaging function result, good cytocompatibility and favorable in vivo effective imaging for CT. Notably, BiNS@NaLnF4 was applied to achieve a satisfactory photon-thermal conversion efficiency (35.3%). Moreover, the special heterostructure barrier achieved increased utilization of electrons/holes, enhancing the generation of reactive oxygen species (ROS) under visible-light irradiation to further expand the therapeutic effect. Dramatically, visible light emission with the up-conversion law was employed to stimulate ROS after irradiation with a 980 nm laser. Simultaneously, the as-prepared BiNS@NaLnF4 can be applied in photothermal/photodynamic therapy (PTT/PDT) investigation for tumor ablation. In summary, the results reveal that BiNS@NaLnF4 is a potential multi-modal theranostic candidate, providing new insights for synergistic theranostics of tumors.


Subject(s)
Photochemotherapy , Theranostic Nanomedicine , Contrast Media , Magnetic Resonance Imaging , Precision Medicine , Tomography, X-Ray Computed
8.
PLoS One ; 13(7): e0200845, 2018.
Article in English | MEDLINE | ID: mdl-30052652

ABSTRACT

The effect of apatinib on the formation of vasculogenic mimicry (VM) was studied in a malignant melanoma cell line. MUM-2B cells cultured in three-dimensional Matrigel were treated with varying concentrations (0, 0.01, 0.05, 0.1, 0.5 µmol/L) of apatinib to test its effect on VM in vitro, followed by MTT proliferation and transwell invasion assays to determine the effect of apatinib on cell proliferation and invasion of MUM-2B cells. In vivo, we used a melanoma cancer model to test the effect of short-term apatinib (100, 200, 300 mg/kg) treatment on VM. Western blotting, immunohistochemistry staining, and CD31-PAS dual staining were performed to assess the expression of VEGFR-2, ERK-1/2, PI3K, and MMP-2, and formation of VM. The results showed apatinib-treated groups formed a lesser number of VM in 3D matrigel, while the cell viability in MTT proliferation assay and the number of migration cells in transwell invasion assay were significantly lower in apatinib-treated groups. In addition, short-term apatinib treatment inhibited angiogenesis, VM formation, and tumor growth in models of melanoma cancer. Mice in apatinib-treated groups showed a markedly reduced expression of VEGFR-2, ERK-1/2, PI3K, and MMP-2. In summary, apatinib could inhibit the expression of VEGFR-2, and downregulate the ERK1/2/PI3K/MMP-2 signaling cascade, which may be one of the underlying mechanisms by which apatinib inhibits angiogenesis and the development of VM in models of melanoma cancer, and restrains the formation of VM by MUM-2B cells. Apatinib shows inhibitory effects on cell proliferation and invasion of MUM-2B cells, which is a close relationship with the VM.


Subject(s)
Antineoplastic Agents/pharmacology , Melanoma/drug therapy , Pyridines/pharmacology , Skin Neoplasms/drug therapy , Animals , Binding Sites , Cell Culture Techniques , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cell Survival , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Humans , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
10.
Drug Deliv ; 24(1): 1410-1418, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28933203

ABSTRACT

The purpose of this study was to prepare endostatin-loaded chitosan nanoparticles (ES-NPs) and evaluate their antitumor effect when combined with paclitaxel (PTX) on Lewis lung carcinoma (LLC) mouse xenografts. ES-NPs were prepared by ionic cross-linking. Characterization of the ES-NPs included size distribution, drug-loading efficiency (DL), and encapsulation efficiency (EE). An in vitro release test was also used to determine the release behavior of the ES-NPs. A subcutaneous LC xenograft model of C57BL/6J mice was established. The mice were randomly divided into six groups: control (0.9% NaCl), ES, PTX, ES-NPs, ES + PTX, and ES-NPs + PTX. The tumor volume was dynamically measured for the duration of the experiment. Immunohistochemistry was performed to determine the Ki-67 and microvascular density (MVD) in each group. Serum vascular endothelial growth factor (VEGF) and ES levels were determined by enzyme-linked immunosorbent assay (ELISA). ES-NPs were successfully synthesized and had suitable size distribution and high EE. The NPs were homogenously spherical and exhibited an ideal release profile in vitro. In vivo, tumor growth was significantly inhibited in the ES-NPs + PTX group. The tumor inhibitory rate was significantly higher in the ES-NPs + PTX group than in the other groups (p < .05). The results of the immunohistochemical assay and ELISA confirmed that ES-NPs combined with PTX had a strong antiangiogenic effect. ES-NPs can overcome the shortcomings of free ES, such as short retention time in circulation, which enhances the antitumor effect of ES. The antitumor effect was more pronounced when treatment included PTX and ES-loaded NPs.


Subject(s)
Carcinoma, Lewis Lung , Nanoparticles , Animals , Cell Line, Tumor , Chitosan , Endostatins , Mice , Mice, Inbred C57BL , Paclitaxel , Vascular Endothelial Growth Factor A
SELECTION OF CITATIONS
SEARCH DETAIL